Axelle Dovonou Animal Models of Parkinson S Disease
Axelle Dovonou Animal Models of Parkinson S Disease
Translational
Translational Neurodegeneration    (2023) 12:36
https://doi.org/10.1186/s40035-023-00368-8                                                                                                      Neurodegeneration
    Abstract
    Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by motor and non-motor symp-
    toms. More than 200 years after its first clinical description, PD remains a serious affliction that affects a growing
    proportion of the population. Prevailing treatments only alleviate symptoms; there is still neither a cure that targets
    the neurodegenerative processes nor therapies that modify the course of the disease. Over the past decades, several
    animal models have been developed to study PD. Although no model precisely recapitulates the pathology, they still
    provide valuable information that contributes to our understanding of the disease and the limitations of our treat-
    ment options. This review comprehensively summarizes the different animal models available for Parkinson’s research,
    with a focus on those induced by drugs, neurotoxins, pesticides, genetic alterations, α-synuclein inoculation, and viral
    vector injections. We highlight their characteristics and ability to reproduce PD-like phenotypes. It is essential to real-
    ize that the strengths and weaknesses of each model and the induction technique at our disposal are determined
    by the research question being asked. Our review, therefore, seeks to better aid researchers by ensuring a concrete
    discernment of classical and novel animal models in PD research.
    Keywords Parkinson’s disease, Animal model, Alpha-synuclein, Transgenic model, Preformed fibril, Mouse
                                        © The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
                                        permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
                                        original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
                                        other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
                                        to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
                                        regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
                                        licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativeco
                                        mmons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Dovonou et al. Translational Neurodegeneration      (2023) 12:36                                                                         Page 2 of 25
 Fig. 1 The hallmarks of Parkinson’s disease (PD). Predominant PD motor symptoms arise from DA neuron loss in the SNpc, and the denervation
 of their axons in the caudate nucleus and putamen, also called the striatum. The cause of PD remains unclear but involves loss of DA neurons, α-syn
 aggregation, mitochondrial dysfunction, autophagy impairments, and neuroinflammation. PD patients additionally exhibit non-motor symptoms.
 The figure was generated using BioRender.
 Fig. 2 Schematic summary of the current known mechanisms that trigger DA neuron death, and the action of different genes and compounds
 used to model PD. The loss of DA neurons could result from impaired protein degradation, mitochondrial dysfunction, α-syn aggregation,
 and neuroinflammation, which can be induced by neurotoxic or genetic alteration. The reduced DA signaling through drug inhibition could
 also lead to a PD-like phenotype. LPS: lipopolysaccharide, DAT: dopamine transporter, VMAT: vesicular monoamine transporter, NM: neuromelanin,
 DA: dopamine. The figure was generated using BioRender.
induces motor impairment as well as memory, cognitive,                     reserpine model is not commonly used to study late-
and emotional deficits [15, 16]. Injection of reserpine par-               stage hallmarks of PD pathology, because it fails to gener-
tially mimics the pathogenesis of PD, developing akinesia                  ate Lewy body-like inclusions [19] and permanent loss of
and rigidity that reflect the clinical features of the dis-                DA neurons. While there are reports of oxidative stress
ease. Furthermore, the work of Carlsson and colleagues                     in the striatum of rodents upon treatment with reserpine,
showed that Levodopa (L-DOPA) can partially rescue                         there is neither indication of mitochondrial or lysosomal
the effect of reserpine administration [10]. In rats, reser-               dysfunction, nor inflammation [20]. However, other than
pine administration produces sexually dimorphic impair-                    memory deficits, reserpine also generates several non-
ments in motor performance, as present in PD [17].                         motor features relevant to the preclinical manifestations
Within weeks, DA neuronal loss in the SNpc and fibers                      of PD, i.e., sleep abnormalities, anxiety, depressive-like
in the dorsal striatum can be seen. However, this pheno-                   behavior, and gastrointestinal dysfunction [21]. For this
type is transient. A partial recovery of nigral DA neurons                 reason, reserpine can be used as a model to study the dis-
occurs 30 days post-injection, rescuing motor deficits                     ease progression and neurochemical features of PD.
[16–18]. It should be noted that no significant loss of DA
axonal innervation in the dorsal striatum is observed in                   Haloperidol
female rats, which could explain the dimorphic impair-                     Haloperidol is a typical antipsychotic that binds post-
ments in motor behavior [17]. Overall, reserpine can                       synaptic dopaminergic D2 receptors [22] (Fig. 2). The
induce PD symptoms in humans and parkinsonian-like                         block of striatal DA transmission results in abnormal
signs in rodents (Table 1). The facility for adjusting dose                downstream firing within the basal ganglia circuits,
concentration and administration allows multiple lev-                      manifesting muscle rigidity and catalepsy [23, 24].
els of studies of PD symptomatology. Nevertheless, the                     Acute administration of haloperidol also reduces the
Dovonou et al. Translational Neurodegeneration          (2023) 12:36                                                                   Page 4 of 25
striatal content of DA, noradrenaline, and serotonin                           of rodents with haloperidol results in significant
[25]. A prolonged administration in mice causes mito-                          increases in the levels of pro-inflammatory cytokines
chondrial complex I (MCI) deficiency in the frontal                            TNF-α and IL-1β in the cortex and striatum compared
cortex, hippocampus, striatum, and midbrain [26]. The                          to control animals [35]. However, no midbrain DA
haloperidol model is used for modelling rigidity, dys-                         neuron degeneration has been reported with haloperi-
kinesia or catalepsy, and for discovering novel antipar-                       dol. Thus, haloperidol may be less relevant for study-
kinsonian agents in rodents [27–30] and non-human                              ing novel neuroprotective or neurorepair strategies for
primates [31–34]. Additionally, chronic treatment                              PD (Table 1).
Drug-induced parkinsonism
Reserpine                   Rodents        Yes          Yes              Yes         NR              NR           NR             Oxidative stress
                                           (SNpc and                                                                             Non-motor phe-
                                           Striatum)                                                                             notype
                                                                                                                                 Transient loss
                                                                                                                                 of DA neurons
                                                                                                                                 Sexual dimorphic
                                                                                                                                 effect
Haloperidol                 Rodents,       Yes          Yes              No          Yes             NR           Yes            Crosses the BBB
                            primates       (Striatum)                                                                            Induces features
                                                                                                                                 of neuroinflam-
                                                                                                                                 mation
                                                                                                                                 No loss of SNpc
                                                                                                                                 DA neurons
Neurotoxic animal models
6-OHDA                      C. elegans*,   Yes          Yes              No          Yes             Yes          Yes            Oxidative stress
                            rodents,       (SNpc and                                                                             Non-motor phe-
                            primates       Striatum)                                                                             notype
                                                                                                                                 Does not cross the
                                                                                                                                 BBB
MPTP                        Rodents and Yes             Yes              Yes         Yes             Yes          Yes            Oxidative stress
                            primates    (SNpc and                                                                                Recreates phases
                                        Striatum)                                                                                of PD
                                                                                                                                 Non-motor phe-
                                                                                                                                 notype
                                                                                                                                 Phenotype vari-
                                                                                                                                 ability in rodents
LPS                         Rodents        Yes          Yes              Yes         Yes             Yes          Yes            Oxidative stress
                                           (SNpc and                                                                             Non-motor phe-
                                           Striatum)                                                                             notype
                                                                                                                                 No α-syn inclu-
                                                                                                                                 sions
                                                                                                                                 Tau pathology
Agrochemical-induced animal models
Rotenone                    C. elegans*,   Yes          Yes              Yes         Yes             Yes          Yes            Oxidative stress
                            Drosophila*    (SNpc and                                                                             Non-motor phe-
                            and rodents    Striatum)                                                                             notype
                                                                                                                                 Inter-individual
                                                                                                                                 variability
Paraquat & Maneb            C. elegans*,   Yes          Yes              Yes         Yes             NR           Yes            Combination
                            Drosophila*    (SNpc)                                                                                increases toxicity
                            and rodents                                                                                          Oxidative stress
                                                                                                                                 Non-motor phe-
                                                                                                                                 notype
                                                                                                                                 Intact striatal fib-
                                                                                                                                 ers in rats
                                                                                                                                 Induces pulmo-
                                                                                                                                 nary fibrosis
Dovonou et al. Translational Neurodegeneration      (2023) 12:36                                                                  Page 5 of 25
Table 1 (continued)
Model                    Species       Dopamine     Motor deficits   α-syn       Mitochondrial   Autophagic   Neuro-         Comments
                                       loss                          pathology   dysfunction     impairment   inflammation
Transgenic models
PRKN & PINK1             C. elegans*   Yes          Yes              N/A         Yes             NR           NR             Lifespan decreases
                         (KO)
                         Drosophila*   Yes          Yes              N/A         Yes             NR           NR             Degeneration
                         (KO)                                                                                                of flight muscles
                                                                                                                             No DA cell loss
                                                                                                                             in PRKN ortholog-
                                                                                                                             KO Drosophila
                         Mice          Yes          Yes              No          Yes             No           Yes            Mitochondrial
                         (KO)          (SNpc                                                                                 dysfunction
                                       and stria-                                                                            Slow degenera-
                                       tum)                                                                                  tion
                         Primates      Yes          Yes              NR          NR              NR           NR             Years to develop
                         (KO)          (SNpc)                                                                                the phenotype
LRRK2                    C. elegans*   Yes          Yes              N/A         Yes             Yes          NR             Progressive loss
                         (OE)                                                                                                of DA neurons
                         Drosophila*   Yes          Yes              N/A         Yes             Yes          NR             Synaptic altera-
                         (OE)                                                                                                tions
                                                                                                                             Tau pathology
                         Mice          Yes          Yes              Yes         Yes             Yes          Yes            Oxidative stress
                         (OE)          (SNpc                                                                                 Synaptic altera-
                                       and Stria-                                                                            tions
                                       tum)                                                                                  Tau pathology
DJ-1                     Mice          Yes          Yes              No          Yes             Yes          NR             May target early
                         (KO)          (SNpc                                                                                 phases of pathol-
                                       and Stria-                                                                            ogy
                                       tum)                                                                                  Age-dependent
                                                                                                                             phenotype
UCH-L1                   Drosophila*   Yes          Yes              N/A         NR              NR           NR             Progressive
                         (KO)                                                                                                age-dependent
                                                                                                                             loss of SNpc DA
                                                                                                                             neurons
                         Mice          Yes          Yes              No          NR              NR           NR             Increased vulner-
                         (OE)          (SNpc                                                                                 ability to α-syn
                                       and Stria-                                                                            Not well charac-
                                       tum)                                                                                  terized
GBA1                     Drosophila*   Yes          Yes              N/A         NR              NR           NR             Increases aggre-
                         (KO/KI)                                                                                             gation in mutant
                                                                                                                             α-syn model
                         Mice          Yes          Yes              Yes         Yes             Yes          Yes            Cognitive deficits
                         (KI)                                                                                                No α-syn aggre-
                                                                                                                             gates in GBA1
                                                                                                                             L444P heterozy-
                                                                                                                             gous mice
Lmx1a/b                  Mice          Yes          Yes              Yes         Yes             Yes          NR             Altered develop-
                         (KO)          (SNpc                                                                                 ment
                                       and Stria-                                                                            KO needs to be
                                       tum)                                                                                  performed
                                                                                                                             on mature animals
Otx2                     Mice          Yes          Yes              N/A         N/A             N/A          N/A            Loss of DA neu-
                         (KO)          (SNpc                                                                                 rons in the VTA
                                       and Stria-                                                                            Altered develop-
                                       tum)                                                                                  ment
                                                                                                                             KO needs to be
                                                                                                                             performed
                                                                                                                             on mature animals
Foxa1/2                  Mice          Yes          Yes              No          N/A             N/A          N/A            Altered develop-
                         (KO)          (SNpc                                                                                 ment
                                       and Stria-                                                                            KO needs to be
                                       tum)                                                                                  performed
                                                                                                                             on mature animals
Dovonou et al. Translational Neurodegeneration      (2023) 12:36                                                                  Page 6 of 25
Table 1 (continued)
Model                    Species       Dopamine     Motor deficits   α-syn       Mitochondrial   Autophagic   Neuro-         Comments
                                       loss                          pathology   dysfunction     impairment   inflammation
Pitx3                    Mice          Yes          Yes              N/A         N/A             N/A          N/A            Altered develop-
                         (KO)          (SNpc                                                                                 ment
                                       and Stria-                                                                            KO needs to be
                                       tum)                                                                                  performed
                                                                                                                             on mature animals
Nurr1& En1               Mice          Yes          Yes              N/A         N/A             N/A          N/A            Total depletion
                         (KD)          (SNpc                                                                                 is not viable
                                       and Stria-                                                                            (Altered develop-
                                       tum)                                                                                  ment)
                                                                                                                             KD needs to be
                                                                                                                             performed
                                                                                                                             on mature animals
c-Rel                    Mice          Yes          Yes              N/A         N/A             N/A          N/A            Altered develop-
                         (KO)          (SNpc                                                                                 ment
                                       and Stria-                                                                            KO needs to be
                                       tum)                                                                                  performed
                                                                                                                             on mature animals
Mitochondrial impairment Mice          No           No               NR          Yes             NR           NR             No phenotypic
                         (POLG)                                                                                              accentuation
                                                                                                                             in DJ-1/PRKN
                                                                                                                             deficient mice
                         Mice          Yes          Yes              NR          Yes             NR           NR             Total reduction
                         (Ndufs2 KO)   (Striatum)                                                                            of MCI activity
                                                                                                                             Mouse death
                                                                                                                             at 5–6 months
                         Mice          Yes          No               Yes         Yes             NR           NR             Partial reduction
                         (Ndufs4 KO)   (Striatum)                                                                            of MCI activity
                                                                                                                             No loss in SNpc
α-syn models
WT α-syn                 Mice          Yes          Yes              Yes         Yes             NR           Yes            No degenera-
                         (M61)         (Striatum)                                                                            tion of SNpc DA
                                                                                                                             neurons
                         C. elegans*   Yes          Yes              Yes         Yes             NR           NR             No endogenous
                         and                                                                                                 α-syn
                         Drosophila*
C-terminally truncated   Mice          Yes          Yes              Yes         NR              NR           NR             Null endogenous
α-syn                    (MI2)         (SNpc                                                                                 α-syn background
                                       and Stria-                                                                            α-syn expression
                                       tum)                                                                                  restricted to
                                                                                                                             DA neurons
E46K human α-syn         Mice          NR           Yes              Yes         NR              NR           Yes            No α-syn in SNpc
                         (M47 line)                                                                                          DA neurons
A30P human α-syn         Mice          NR           Yes              Yes         Yes             Yes          NR             Around a year
                                                                                                                             to develop strong
                                                                                                                             phenotype
                                                                                                                             No degenera-
                                                                                                                             tion of SNpc DA
                                                                                                                             neurons
                         C. elegans*   Yes          Yes              Yes         Yes             NR           NR             Lower α -syn
                         and                                                                                                 accumulation
                         Drosophila*                                                                                         than α-syn WT
                                                                                                                             Lower α -syn
                                                                                                                             accumulation
                                                                                                                             than α-synA53T
A53T human α-syn         Mice          NR           Yes              Yes         Yes             Yes          Yes            α-syn inclusions
                         (M83 line)                                                                                          in non-PD-related
                                                                                                                             areas
                         C. elegans*   Yes          Yes              Yes         Yes             NR           NR             No endogenous
                         and                                                                                                 α-syn
                         Drosophila*
Dovonou et al. Translational Neurodegeneration             (2023) 12:36                                                                                   Page 7 of 25
Table 1 (continued)
Model                          Species       Dopamine       Motor deficits     α-syn          Mitochondrial      Autophagic       Neuro-            Comments
                                             loss                              pathology      dysfunction        impairment       inflammation
idiopathic PD, where neuronal degeneration occurs over          Gram-negative bacteria and is well-known for its pro-
the years. Despite its limitations, it remains an excellent     inflammatory properties. LPS binds the toll-like recep-
tool for studying anti-PD drugs, L-DOPA-induced dyski-          tor 4 mainly expressed in microglia [65]. The use of LPS
nesia, or cell transplantation therapy.                         was initially intended for systemic inflammation models
                                                                and the discovery of novel therapies in the treatment of
1‑Methyl‑4‑phenyl‑1,2,3,6‑tetrahydropyridine (MPTP)             acute inflammation [66, 67]. However, in 2001, a clinical
First discovered in humans, this neurotoxin soon became         case report showed that a 22-year-old laboratory worker
a vital research tool in the early 1980s. Mistaken for syn-     developed PD syndrome after accidental exposure to
thetic heroin, MPTP produced clinical and pathological          Salmonella minnesota LPS through an open wound.
PD features in young drug addicts [56]. After systemic          Positron emission tomography confirmed that the sub-
administration, MPTP rapidly crosses the BBB and is             ject also suffered from a dopaminergic neuronal loss in
metabolized to MPDP+ (1-methyl-4-phenyl-2,3-dihy-               the SNpc [68]. Furthermore, two studies published ear-
dropyridinium) by astrocytes [57]. The active toxic com-        lier showed that injecting LPS into the SNpc of rats leads
pound, 1-methyl-4-phenylpyridinium (MPP+), quickly              to neurodegeneration of nigral DA neurons [69, 70]. The
enters DA neurons through DAT, where it binds to the            neuronal loss is permanent and selective to DA neurons
VMAT and translocates to synaptosomal vesicles or               [71]. Since then, several LPS-induced models of PD have
remains within the mitochondria and cytosol (Fig. 2).           been developed, but their phenotypes greatly depend on
MPP + leads to an acute deficit in ATP formation and            the administration route [65, 72]. LPS can be delivered
ROS production, via binding to MCI, and ends in neu-            systemically or locally to the striatum, pallidum, or SNpc
ronal death [58]. Furthermore, MPTP administration              through intracerebral injections or intranasally.
drives microglial activation and cytokine release [59]. In         While a few studies have used mice, LPS is more com-
humans and non-human primates, MPTP can replicate               monly used in rats [73–75]. Intrastriatal and intranigral
almost all the motor impairments of PD, including trem-         injection of LPS in rats results in severe neurodegenera-
ors, rigidity, slowness of movement, postural instability,      tion of DA neurons from the SNpc with reduced striatal
and freezing [60]. The loss of SNpc DA neurons can also         DA and motor impairments within a month [76–83].
be accompanied by an almost complete depletion in DA            Reduced MCI and II can be observed in nigral neurons
and associated metabolites in the putamen [61]. It must         [78]. A similar phenotype is seen when injecting LPS
be noted that this model is highly variable depending on        into the pallidum of rats, but motor impairments appear
the neurotoxin dosage, and the species and strains used.        transient and return to normal after a few weeks [84].
Unlike primates, rodents (and variable strains of mice)         It should be noted that using older rats can increase
are less sensitive to MPTP toxicity and require multiple        the severity of the phenotype [78, 84]. Furthermore, it
doses over several days to induce neuronal degeneration.        has been reported that LPS increases the expression of
Thus, an appropriate administration schedule is needed          α-syn in the brain [78, 85], the amount of oligomerized
[62]. While mice treated with MPTP do not develop per-          α-syn [86], and the expression of cytokines and activated
sistent and progressive motor dysfunctions [60], they are       microglia in both the SNpc and the striatum [78, 82].
arguably a viable choice for studying neuroanatomical           Overall, local delivery of LPS to the brain generates fea-
and neurochemical alterations. In the SNpc and ventral          tures relevant to PD and can thus be used to clarify the
tegmental area (VTA) of treated mice, MPTP causes the           role of neuroinflammation in SNpc DA neuron degenera-
loss of more than half of all DA neurons. Recent stud-          tion. However, the pathology is limited to the injection
ies show that the administration of MPTP can increase           site.
the total and the phosphorylated α-syn as well as tubu-            Systemic injection of LPS in mice through intraperito-
lin-associated unit (Tau) in the hippocampus and SNpc           neal injection induces loss of DA neurons, motor deficits,
[63]. Hence, MPTP is widely used as a model to study the        and α-syn accumulation [85, 87, 88]. Autophagic impair-
molecular and neuropathological events in PD, especially        ment has also been described in nigral neurons with
in non-human primates (Table 1).                                this route of administration [89]. It has been reported
                                                                that there may be a delay in the appearance of the phe-
Lipopolysaccharide (LPS)‑induced neuroinflammation              notype depending on the recurrence of the dose injected
In PD patients, a dense population of reactive micro-           [72]. After a single intraperitoneal injection of LPS in
glia and astrocytes can be found in the brain, accom-           mice, neurodegeneration of DA neurons can take sev-
panied by an increased amount of pro-inflammatory               eral months [87, 88]; this delay can be reduced to 1 week
cytokines, suggesting that neuroinflammation could play         by performing daily injections of LPS [85]. Recently, the
a key role in the pathogenesis [64]. Neuroinflammation          administration of LPS through the nasal route in mice
can be modeled using LPS, an endotoxin produced by              has gained popularity. A chronic administration via the
Dovonou et al. Translational Neurodegeneration   (2023) 12:36                                                    Page 9 of 25
PRKN and PINK1                                                    primates has been generated. This model requires spe-
Of all the familial forms of PD, PRKN is the most fre-            cialized expertise and installations to maintain these ani-
quently mutated gene accounting for about 50% of early-           mals, which may exceed the technical capacities of many
onset genetic cases, followed by mutations in PINK1,              research teams.
which represent up to 8% of early-onset familial forms
of PD [127]. PRKN and PINK1 are both involved in the              LRRK2
mitochondrial stress response and are believed to act             Mutations in the LRRK2 gene are dominant and account
along the same pathway. Indeed, mitochondrial stress              for around 4% of familial cases of PD [139]. LRRK2 is a
leads to the accumulation of PINK1 within mitochon-               protein containing both a kinase and a GTPase domain.
dria and initiates autophagy of damaged mitochondria              In addition, this gene interacts with many proteins,
through the recruitment of PRKN [129] (Fig. 2). PRKN              regulating various cellular functions (Fig. 2), including
and PINK1 mutations are recessive and generally lead to           autophagy, mitochondrial functions, and vesicular traf-
a loss of function. Interestingly, almost all clinical cases of   ficking [140]. Most LRRK2 mutations, such as G2019S
PRKN-induced parkinsonism lack Lewy body pathology                and R1441C/G, affect one of the enzymatic domains.
[130].                                                            Hyperactivity of LRRK2 kinase has also been observed
   In C. elegans and Drosophila, genetic deletion of the          in idiopathic cases [141]. While DA neuron loss from
ortholog form of human PRKN and PINK1 results in                  the SNpc is one characteristic of parkinsonian patients
typical Parkinson-like phenotypes. In C. elegans, these           carrying a LRRK2 mutation, approximately 21%–54%
mutations are associated with motor impairment, mito-             of these clinical cases do not show apparent Lewy bod-
chondrial dysfunction, and loss of DA neurons [131,               ies [149, 150], suggesting that this mutation could induce
132]. In Drosophila, PINK1 ortholog deletions lead to the         neurodegeneration through another factor than synucle-
development of similar features, with the addition of the         inopathy. Some evidence indicates that the toxic effect
degeneration of muscles [133]. PRKN ortholog-deficient            might be mediated through the protein Tau, since about
Drosophila have a comparable phenotype, except for                79% of PD patients carrying a LRRK2 mutation show
the absence of DA neuron loss [134]. These models are             some features of Tau pathology [142]. In C. elegans and
particularly suitable for fast screening approaches when          Drosophila, overexpression of human wild-type (WT) or
studying PRKN/PINK1 regulatory mechanisms involved                G2019S and mutated LRRK2 causes motor dysfunctions
in the pathogenicity. In mice, the genetic deletion of            and progressive degeneration of DA neurons [144–148].
both PINK1 and PRKN leads to mitochondrial dysfunc-               LRRK2 G2019S overexpression in both species results
tion and neuroinflammation without affecting the basal            in mitochondrial and autophagic dysfunction [144, 146,
mitophagy. This model does not appear to exhibit α-syn            147, 149]. In Drosophila, overexpression of the G2019S
aggregates, behavioral abnormalities, reduced striatal            mutated form of LRRK2 induces a loss of dendritic arbo-
DA levels, or loss of nigral DA neurons [135, 136]. How-          rization by hyperphosphorylation of endogenous Tau.
ever, it is possible that neurodegeneration only occurs in        Furthermore, overexpression of the Drosophila ortholog
older mice. In fact, a recent study showed that hind limb         of WT LRRK2, Lrrk, has been seen to increase the neu-
defects and loss of DA neurons in the SNpc can be seen in         rotoxicity of pathological Tau when the human mutated
2-year-old PRKN knock-out (KO) mice [137]. Thus, mice             R406W form of Tau is expressed [148]. The Drosophila
lacking PINK1 or PRKN might be used to better under-              model can therefore be useful in understanding the inter-
stand the role and the involvement of these genes in vivo.        action of LRRK2 and Tau pathology. Moreover, as C.
Nevertheless, the time required for these mice to develop         elegans and Drosophila do not naturally express α-syn,
neurodegeneration could be a major limiting factor for            the function of LRRK2 can additionally be studied in the
using this model in neuroprotective studies. Recently, a          absence of endogenous α-syn.
non-human primate model with a genetic deletion of                   In mice, G2019S LRRK2 overexpression and Knock-
PINK1 was generated. This model presents behavioral               In (KI) models generate synaptic alterations, impaired
abnormalities at 1.5 years of age and DA neuron degen-            DA transmission, behavioral abnormalities and
eration in the SNpc at 3 years of age, without changes in         increased Tau phosphorylation [150–156]. These
mitochondrial morphology [138]. It is still unknown if            rodent models usually lack degeneration of DA neu-
these animals develop Lewy body-like inclusions. While            rons in the SNpc, unless the mouse line overexpresses
the loss of PINK1 more closely reflects parkinsonian              G2019S LRRK2 under the neuron-specific platelet-
phenotypes, this model is not yet fully characterized.            derived growth factor beta (PDGF-β) promoter. These
Further studies are needed to assess its importance as a          transgenic mice show striatal denervation, DA neuron
PD animal model (Table 1). Although promising and rel-            degeneration, and increased phosphorylated Tau pro-
evant to PD, no PRKN-transgenic model in non-human                tein in the SNpc at 16 months of age [157]. Features of
Dovonou et al. Translational Neurodegeneration   (2023) 12:36                                                    Page 11 of 25
neuroinflammation, as well as autophagic and mito-              DA axonal denervation in the striatum, DA neuron loss
chondrial abnormalities are also present despite the            in the SNpc, and motor impairments after 2 weeks [165].
absence of synucleinopathies [158, 159]. More recently,         The MPTP treatment accelerates the phenotype seen in
neurodegeneration has been reported in a mouse line             KO models alone by increasing the oxidative stress bur-
expressing G2019S LRRK2 in DA neurons under the                 den [163] (Table 1).
TH promoter. The alteration drives a progressive loss
of DA neurons in the SNpc by 15 months of age, with             UCH‑L1
additional neuronal loss observed in the olfactory bulb         UCH-L1 is a deubiquitinating enzyme believed to be one
and locus coeruleus. By 24 months of age, this pheno-           of the most abundant proteins in neurons, accounting for
type is accompanied by reduced synaptic vesicles in the         about 1%–5% of the total amount of proteins in the brain
DA neurons, motor deficits, and an increased amount             [172]. The first association between the UCH-L1 gene
of phosphorylated α-syn [160] (Table 1). Overall,               and PD was revealed in 1998, following the identification
LRRK2 mutant mice might be used to study the patho-             of the autosomal mutation I93M in the pedigree of a Ger-
genesis of PD and to identify neuroprotective therapies         man family [173]. Although this mutation is rare, UCH-
aimed at inhibiting its kinase activity or its downstream       L1 localizes with Lewy bodies in some sporadic cases of
targets.                                                        PD [174, 175]. While UCH-L1 is involved in the clear-
                                                                ance of proteins via the ubiquitin-proteasome system,
                                                                a recent study also suggests that UCH-L1 can modulate
DJ‑1                                                            mitochondrial functions by regulating the expression of
First discovered as a novel oncogene in 1997 [161],             Mitofusin-2 [176] (Fig. 2). To date, there are relatively few
the DJ-1 gene encodes a protein of the peptidase C56            transgenic animal models for UCH-L1. In mice, the dele-
family. It is also known for its function against oxida-        tion of UCH-L1 induces axonal degeneration in sensory
tive stress [162, 163] via its direct interaction with          and motor nerve terminals [177–179] without affect-
mitochondria [164] and DA neurons in the SNpc,                  ing DA nigral neurons [175]. The only transgenic mouse
which drive the nigrostriatal motor pathway inhibi-             model that affects the DA neurons of the SNpc is the
tion (Fig. 2) [165]. Deficiency of DJ-1 has been identi-        UCH-L1 I93M mice, which overexpress I93M mutated
fied as a causative factor in familial PD with recessive        UCH–L1 under the control of the PDGF-β neuron-spe-
inheritance [166], where it downregulates the level             cific promoter [180]. Although these mice may be useful
of lysosomal 70 kDa heat-shock cognate protein. By              for studies aimed at discovering neuroprotective thera-
simultaneously inhibiting the activation of chaperone-          pies for PD by targeting UCH-L1 activity, this approach
mediated autophagy, DJ-1 deficiency may augment                 is limited because UCH-L1 I93M transgenic mice lack
α-syn aggregation [167] and mitochondrial abnor-                α-syn inclusions. Furthermore, the degeneration of
malities [168] in PD. Although DJ-1 KO mice exhibit             DA neurons and motor deficits can take more than 20
alterations in DA metabolism, the KO alone is not suf-          months to develop [180]. To accelerate the degeneration
ficient to induce loss of DA neurons and PD-like signs          of SNpc DA neurons, this model has been previously
in young mice. However, in older mice, there are sig-           combined with viral α-syn overexpression [175]. More
nificant DA neuronal losses and motor deficits [169].           recently, knock-down (KD) of the UCH-L1 ortholog gene
A similar age-dependent difference is also visible in           (dUCH) in a Drosophila PD model was created using the
the Drosophila model with two existing orthologs for            GAL4-UAS system. This model exhibits motor altera-
DJ-1 [170]. While the major limitation of this model            tions, degeneration of DA neurons, and a reduction in
lies within the prolonged time required for a phenotype         the level of DA in the brain [181], thus making this model
to appear, it remains promising, as ageing seems to be          appropriate for fast screening approaches to identify
a crucial factor in progressive neurodegenerative dis-          UCH-L1 interactors (Table 1).
eases like Parkinson’s. Because PD similarly evolves in
older adults, this progressively developing model may
be suitable for targeting the earlier phases of the phe-        GBA1
notype induction and its subsequent manifestation.              Mutations in GBA1 are among the most common genetic
   Recent studies have used MPTP hybrid models that             risk factors for PD and Lewy body dementia [182, 183].
take advantage of the usual role DJ-1 has as a negative         The GBA1 gene is located on chromosome 1q21 and
regulator of the inflammatory response, alleviating neu-        encodes the lysosomal glucocerebrosidase enzyme
roinflammation, and playing an essential role in neurode-       (GCase) [184]. Thus, GBA1 mutations induce a defi-
generation [171]. DJ-1 KO mice tend to present PD-like          ciency of GCase activity that leads to an accumulation
pathology after MPTP treatment, resulting in accelerated        of glycosphingolipids [185]. Clinical, epidemiological,
Dovonou et al. Translational Neurodegeneration   (2023) 12:36                                                 Page 12 of 25
and experimental studies have confirmed a connection            features lead to DA neuron death and motor phenotypes,
between PD and Gaucher disease, a prevailing autoso-            making them an appealing tool for studying PD (Table 1).
mal recessive lysosomal storage disorder [186]. Recently,
GBA1-derived PD clinical cases have shown impaired DA           Transcription factors
neurotransmission early in the pathogenesis, although           During the development of midbrain DA neurons, tran-
the underlying mechanisms are still not well understood.        scription factors play key roles in specifying and dif-
So far, approximately 130 different GBA1 mutations have         ferentiating neural progenitors. Interestingly, most of
been observed in patients with PD. The most frequent            these transcription factors remain expressed in the adult
cases with declined GCase activity are the L444P and            brain [199, 200]. Indeed, Lmx1a/b, Otx2, Foxa1/2, Pitx3,
N370S GBA1 mutations [185, 187, 188]. Recent stud-              Nurr1, and En1/2 are present throughout development
ies have focused on the association between mutated             and are required for the survival of midbrain DA neu-
GBA1 and PD, highlighting the possibility that lysosomal        rons [201–204]. Genome-wide association studies have
impairment and altered GCase activity could facilitate          revealed genes that may contribute to PD [205]. Among
the proliferation of α-syn aggregates [188–190]. Today,         them, genetic variants of human transcription factors
several animal models of GBA1 have been created [182].          such as Lmx1a and Lmx1b were found [204, 206–208],
GBA1-associated PD mouse models combine a GBA1                  with the expression of Lmx1b being notably decreased in
mutation with overexpression of α-syn mutations to gen-         the DA neurons of PD patients [209]. Lmx1a and Lmx1b
erate α-syn aggregation and pronounced PD-like signs.           are LIM homeodomain transcription factors essential for
Additionally, different KI models carry the human L444P         the development of midbrain DA neurons [210–213] and
GBA1 gene [191]. GBA1 L444P homozygous mice show                are still expressed in post-mitotic midbrain DA neurons
an increase of α-syn accumulation in the striatum and           [214–216]. Conditional deletion of Lmx1a and Lmx1b
astrogliosis at 1 year of age [192]. GBA1 L444P heterozy-       genes in mouse DA neurons induces progressive neuro-
gous mice develop mitochondrial dysfunction, increased          degeneration in the SNpc and VTA [203, 217]. Lmx1a
ROS production, impaired neuronal autophagic degra-             and Lmx1b are particularly important in regulating mito-
dation [193], and higher levels of α-syn [194], but fail to     chondrial [217] and autophagic-lysosomal functions
develop α-syn aggregates even by 24 months of age [194].        [209]. Moreover, loss of Lmx1a and Lmx1b leads to α-syn
Unlike L444P, heterozygosity for N370S does not affect          pathology with the accumulation of α-syn in both DA
phenotype progression in A30P-SNCA mice [193], and              axons and cell bodies [217]. Specific deletion of Lmx1a
N370S homozygote mice die at birth. Injection of adeno-         and Lmx1b in adult DA neurons also leads to progres-
associated virus (AAV) expressing N370S GBA1 in the             sive loss of DA neurons, α-syn pathology, and motor
striatum of A53T-SNCA mice results in increased α-syn           impairments [209, 217]. Depletion of Otx2, Foxa1/2, and
protein, defects in mitophagy, and abnormal autophagy           Pitx3 leads to a reduction of midbrain DA neurons with
[191, 193]. GBA1-associated PD models can also be               motor deficits [218–223]. While heterozygous KO mice
found in Drosophila and C. elegans. In these PD-GBA             for Nurr1 and En1 develop progressive loss of DA neu-
Drosophila models, an increased aggregation of mutant           rons with motor deficits [224, 225], total depletion of
α-syn A53T is observed, causing enhanced DA neuronal            Nurr1 [224, 226] and En1 [227] in midbrain DA neurons
loss and exacerbating the motor and non-motor pheno-            is embryonically lethal. Lastly, KO of the transcription
types [195]. Drosophila expressing the mutant human             factor c-Rel can induce α-syn aggregation in the SNpc,
N370S and L444P variants exhibit a significant decrease         midbrain DA neuronal loss, and motor deficits [228]
in GCase activity and present parkinsonian features with        (Table 1). Overall, several signs of PD can be reproduced
DA cell death, defective locomotion, and a shorter lifes-       by modulating the expression of DA transcription fac-
pan [196]. Overall, fly models expressing human WT              tors; however, the phenotype is restricted to DA neurons.
or N370S and L444P GBA1 provide an interesting tool             Most of these models also require multiple crosses to
to assess the contribution of defective GCase function          obtain a conditional deletion in adult mice, which could
to PD development. In C. elegans expressing a human             thus represent a limiting factor.
α-syn, KD of GCase ortholog, C33C12.8, increases aggre-
gation of α-syn [197], similar to what has been described       Mitochondrial impairment
in other animal models [198]. In sum, GBA1-associated           Several lines of evidence have previously linked mito-
models of PD provide insight into the pathogenesis by           chondrial dysfunction to PD pathogenesis [229, 230].
replicating many hallmarks of the disease. GBA1 models          This is supported by a large set of PD-associated genes
show increased α-syn accumulation, disruption of lyso-          that are involved in mitochondrial function [231, 232].
somal homeostasis, exacerbated endoplasmic reticulum            Furthermore, α-syn and neurotoxins linked to PD,
stress, and mitochondrial dysfunction. These cellular           including MPTP and rotenone, are known to inhibit
Dovonou et al. Translational Neurodegeneration   (2023) 12:36                                                  Page 13 of 25
MCI activity [233]. MCI deficiency was also reported            eight different identified missense mutations of SNCA
in DA neurons of the SNpc for human PD post-mortem              (A53T/E/V, A30P/G, E46K, H50Q and G51D) [244],
brains [234]. One of the first transgenic mouse models          gene duplication, and triplication have been linked to the
of PD to induce mitochondrial impairment in DA neu-             accumulation of pathologic α-syn in the brain [245]. This
rons was created by crossing DJ-1-deficient mice with a         new knowledge gave rise to several animal models mim-
mouse strain encoding a mutated mitochondrial poly-             icking α-syn pathology. Transgenic models expressing
merase gamma, resulting in an accelerated accumulation          WT, A53T, A30P, and E46K SNCA mutations have been
of mitochondrial DNA errors. Even at 1 year of age, these       utilized to recapitulate PD pathology in vivo. However,
mice do not exhibit behavioral abnormalities, degenera-         to date, there are no reported studies on A53E, A53V,
tion of nigrostriatal DA neurons, or evidence of neuroin-       A30G, H50Q, or G51D transgenic models, which could
flammation [235]. A similar phenotype has recently been         be of interest for future investigations.
observed in mice lacking PRKN [236].
   In mice, conditional deletion of the Ndufs4 gene (which      WT and truncated human α‑syn
encodes a subunit of MCI) in DA neurons results in loss         As one of the first transgenic animal models of α-syn,
of striatal DA at 9 months, followed by increased α-syn         the mouse line 61 (M61) is generated by overexpressing
phosphorylation in DA neurons at 24 months of age.              human WT α-syn using the neuron-specific Thymocyte
However, deletion of Ndufs4 in DA neurons does not              differentiation antigen 1 (Thy1) promoter [246]. Mito-
cause motor deficits or DA neuron degeneration in the           chondrial dysfunction and neuroinflammation have been
SNpc [237]. One possible explanation for this phenotype         described in these mice [247, 248]. Moreover, motor defi-
is that Ndufs4 KO results in about 65% reduction of MCI         cits and α-syn inclusions appear within a few months of
activity [237] and DA neurons could compensate for the          age, although nigral DA neurons are only mildly affected.
lack of oxidative phosphorylation [238]. Recently, another      In fact, while a decreased amount of striatal DA and axonal
model of MCI disruption by deletion of the essential MCI        denervation are observed, no degeneration of DA neurons
subunit Ndufs2 in DA neurons was generated. Com-                in the SNpc is reported [246, 249]. M61 mice have thus
pared to the Ndufs4 KO model, the KO of Ndufs2 almost           been used to test candidate drugs targeting α-syn aggrega-
completely disrupts the oxidative phosphorylation activ-        tion and axonal denervation. The MI2 mouse line express-
ity in DA neurons. Deletion of Ndufs2 induces loss of           ing C-terminal-truncated human α-syn under the TH
DA axons in the dorsal striatum by 1 month of age, fol-         promoter shows α-syn aggregates in DA neurons by 1.5
lowed by motor deficits after 1 to 2 months. These motor        months of age. By 9 months, motor impairments appear;
deficits are accompanied by a decrease in the number of         by 12 months, a significant loss of DA neurons in the SNpc
cells expressing TH in the SNpc. However, despite the           can be seen [250]. Additionally, because MI2 mice pos-
decrease in TH expression, there is no loss of dopamin-         sess a null endogenous α-syn background, human α-syn
ergic cell bodies [238]. No α-syn aggregates have been          seeding is facilitated. However, since the α-syn expression
observed in this model, but it might be because that            is restricted to DA neurons, the MI2 mouse line does not
these mice die at 5–6 months of age before developing           recapitulate synucleinopathies in other brain regions.
α-synucleinopathies [238]. Additional characterization
of neuroinflammatory and autophagic features is still           E46K human α‑syn
needed. However, the Ndufs2 KO mouse model remains              Identified within a Spanish family with autosomal domi-
of interest in the study of neuroprotective therapies aim-      nant parkinsonism and dementia, the cortical and sub-
ing to rescue oxidative phosphorylation or MCI activity.        cortical Lewy body pathology found was attributed to
                                                                a shared nonconservative heterozygous E46K mutation
α‑syn models                                                    within α-syn [251]. While the mutant mouse PD models
The α-syn protein, encoded by SNCA, has a central role          created based on this mutation are still recent and war-
in PD, although its physiological function is not yet           rant further research, this initiative can provide insight
fully understood. Usually clustering into monomers and          into familial PD and synucleinopathies. Transgenic
tetramers [239, 240], α-syn assembles into higher-order         mouse lines carrying the PrP (Prion protein)-driven
structures under pathological conditions. These oligom-         E46K mutation have been generated. The mouse line 47
ers, protofibrils, and fibrils form insoluble aggregates        (M47) shows higher levels of α-syn transgene expres-
that are toxic to neurons [241]. As a principal constitu-       sion, with motor impairments starting at 15–16 months
ent of Lewy bodies [242], these aggregates can not only         of age. These mice develop α-syn inclusions in the spinal
alter the integrity of the cell membrane, but may also dis-     cord, brainstem, deep cerebellar nuclei, motor cortex,
rupt the functions of mitochondria, the Golgi apparatus,        and most of the thalamus. The inclusions and affected
and the endoplasmic reticulum [243] (Fig. 2). At least          areas are accompanied by phosphorylation of α-syn,
Dovonou et al. Translational Neurodegeneration   (2023) 12:36                                                   Page 14 of 25
significant astrocytic gliosis, elevated reactive micro-        phenotype with evidence of dystrophic neurites. They
glia, and Tau inclusions [252]. DA neurons of the SNpc          present increased phosphorylated α-syn and aggregation,
remain spared of α-syn inclusions. These results may            similar to patients [264–267]. These inclusions become
corroborate those observed in rat models with the same          apparent between 8 to 16 months of age in homozygous
mutation, where no loss of DA neurons or striatal termi-        M83 mice, resulting in motor impairments. Like M47
nals was reported [253].                                        mice, the highest density of these inclusions is localized
                                                                in the spinal cord, brain stem, deep cerebellar nuclei,
A30P α‑syn                                                      white matter, and some thalamic regions. An identical
The A30P α-syn mutation was first detected in a German          inclusion profile arises in hemizygous M83 mice between
family [254]. The few existing transgenic mouse strains         22 and 28 months of age when animals spontaneously
that model the A30P mutation include overexpression             develop motor dysfunction [264]. No loss of DA neurons
of the human A30P α-syn gene under the Prp promoter             has been reported, and there is no alteration of striatal
(Prnp-A30P) [255] or the Thy1 promoter (Thy1-A30P)              DA at 12 months of age in M83 homozygous mice [268].
[256], and a KI mouse model with an introduced A30P             Altogether, this mutation owes its pathological nature to
point mutation in the WT gene [255]. All these A30P             its tendency to form α-syn inclusions, much like in the
models show an increase and accumulation of α-syn in            clinical setting. While the location of pathological lesions
neuronal cell bodies with age. However, it is most robust       in mice does not precisely mirror the human condition,
in the Prnp-A30P mice. For Thy1-A30P α-syn transgenic           they do share many similarities, such as neuroinflam-
mice, abnormal α-syn protein is observed in several brain       mation, as well as autophagic and mitochondrial dys-
regions like the superior colliculus or the cerebellum, but     function [269–272]. This human A53T mutation model
α-syn pathology is spared in the striatum and SNpc [255].       is proposed to be a valuable asset in screening potential
Mitochondrial oxidative stress and autophagy defects            therapeutics that inhibit or reverse α-syn aggregate for-
are also reported in A30P models [257]. Only the neu-           mation or spreading.
ronal cell loss in the SNpc does not seem to be consist-
ently detectable. While Thy1-A30P mice show reduced             α‑syn preformed fibrils (PFFs)
TH levels in the SNpc at 8 and 11 months [256], the             PFFs are first generated from monomeric recombinant
loss of DA is not visible in the strains [255, 258]. Loco-      α-syn. When fragmented by sonication, PFFs can phos-
motor disabilities and α-syn aggregation seen in rodent         phorylate endogenous α-syn into its pathological state
models are similarly observed in flies overexpressing the       [273]. In mice, α-syn spreading is enhanced with PFFs
A30P mutant form of α-syn. Flies also exhibit selective         generated from brain homogenates of aged sympto-
loss of DA neurons when A30P is expressed in all neu-           matic α-syn mice or human samples, or recombinant
rons [259, 260]. In C. elegans, A30P α-syn models are           synthetic α-syn fibrils (Fig. 2). In WT mice, injection of
mild. Compared to control animals, worms expressing             mouse PFFs in the dorsal striatum induces hyperphos-
A30P α-syn in muscles have modest reduction of move-            phorylation of α-syn within 30 days [274]. Degeneration
ment speed and increased rate of paralysis [261]. In a C.       of DA striatal fibers and cell bodies in the SNpc, as well
elegans model of A30P under the control of the DAT-1            as motor impairments, occurs 6 months post-injection
(C. elegans DAT) promoter, the expression is restricted         [274]. Whole-brain homogenate extracts of parkinsonian
to DA neurons. The worms exhibit DA neuron-specific             symptomatic M83 mice can be injected into the M83
dysfunction caused by accumulation of α-syn but do              mice at birth to accelerate the development of the phe-
not show significant neurodegeneration compared to              notype and α-syn aggregation [275–277]. There is also a
controls, even at 15 days of age [262]. Overall, the A30P       robust inflammatory response before end-stage paraly-
models show mild phenotypes. The progressive and rela-          sis occurs by 8 to 16 months in homozygous mice and by
tively long development of pathological abnormalities,          22 to 28 months in heterozygous mice [278]. Similarly,
such as α-syn accumulation and motor disorders, may             extracts from post-mortem PD brain tissues have previ-
provide an interesting approach to study the early stages       ously been used in the intranigral and intrastriatal inocu-
of PD.                                                          lation of WT mice and macaque monkeys. The fractions
                                                                enriched with nigral Lewy bodies have demonstrated
A53T human α‑syn                                                the ability to induce pathogenic effects, such as progres-
The A53T mutation in the α-syn gene was initially iden-         sive nigrostriatal neurodegeneration and the accumu-
tified in Italian and Greek pedigrees [263]. The PrP-           lation of pathological α-syn, which cannot be achieved
driven mouse line 83 (M83) was the first to be genetically      by non-Lewy body fractions or vehicle buffers [279]. A
engineered to overexpress human A53T α-syn. M83                 new alternative created with the bacterial artificial chro-
mice show a mid-to-late onset of neurodegenerative              mosome plasmid consists of A53T overexpressing mice
Dovonou et al. Translational Neurodegeneration   (2023) 12:36                                                   Page 15 of 25
injected with recombinant synthetic α-syn. This hybrid          interest [301]. This technique thus makes it feasible to
model produces phosphorylated α-syn inclusions and              study the possible transmission of α-syn pathology from
striatal DA denervation within 2 weeks post-injection           the periphery to the central nervous system [302]. In gen-
[280]. Within 1 month, there is 25% DA neuronal loss            eral, α-syn overexpression in the SNpc results in the pro-
in the SNpc. Motor impairments were reported at 2               gressive DA neuron loss, and DA projection decrease in
months post-injection [280], but they can be expected           the striatum within weeks to months, and models early
to appear earlier since DA denervation appears before           and late hallmarks of PD pathogenesis, depending on the
this time point [277]. In cell-based models, PFFs were          model organism [300, 303–305]. However, studies have
shown to additionally produce autophagic and mito-              yielded variable results, showing a range from slight to
chondrial dysfunction [281, 282]. Lastly, recent studies        substantial loss of DA neurons. Several factors, includ-
in 26-month-old non-human primates have shown that              ing virus titer or serotype, age, sex, strain, or species of
PFFs are also sufficient to drive phosphorylated α-syn          the animal, can strongly influence the outcomes obtained
accumulation and DA neuronal loss by 3 months post-             (see [306, 307] for reviews). In rodents, viral α-syn over-
injection [273]. However, the injection site of the PFFs is     expression results in an approximate 40% DA cell loss
consequential in further characterizing the α-syn seeding       in the SNpc, accompanied by motor deficits at 8 weeks
and spreading [274, 277, 283]. We cannot overlook that          post-injection [308]. Less commonly used, non-human
α-syn transmission also occurs in the periphery and that        primates receiving intranigral AAV injections of WT or
α-synucleinopathy in the brain may develop via the gut-         A53T α-syn show a ~ 30% loss of nigral DA neurons 8
to-brain axis [284]. Inoculation of the rodent gut with         weeks post-injection. Like rodents, non-human primates
PFFs results in midbrain DA neuron loss, α-syn histopa-         also develop motor deficits with a progressive deteriora-
thology, and motor defects in a temporal manner [285–           tion of motor coordination after 16 weeks, albeit with a
287]. Nevertheless, despite differences from the human          greater variability [309]. Non-human primates enable a
pathology, PFFs can reproduce specific aspects or stages        more extended period of studies on the effects of α-syn
of α-syn pathology, as well as neurodegenerative and            injections than their rodent counterparts [310, 311].
neuroinflammatory hallmarks of PD [288]. Overall, both          Moreover, monkeys were found to be more vulnerable to
in vitro and in vivo, fibril formation and aggregation are      neurodegeneration and pathologies induced by injections
not synonymous with pathological aggregation and LB             with viral vectors that encode A53T α-syn [311]. Overex-
formation. Therefore, strict quality control must be taken      pression of α-syn in the midbrain typically leads to mito-
so that the pathogenicity of the PFFs used ensures repro-       chondrial dysfunction [312], impaired autophagy [313],
ducible results [284, 289, 290].                                Lewy body-like structures [300, 314], and an early and
                                                                persistent inflammatory response [315–318]. Recently,
Non‑mammalian models of α‑syn                                   a light-inducible α-syn aggregation system (LIPA) was
Non-mammalian models, including those based on                  designed to regulate spatiotemporal protein aggregation
C. elegans and D. melanogaster, do not produce α-syn            [319]. This system could control α-syn aggregation into
endogenously. In C. elegans [291–293] and Drosophila            insoluble deposits that mimic key features of Lewy bod-
[294–296], the overexpression of human WT and A53T              ies. LIPA is encoded in a viral vector and injected into
α-syn in an ubiquitous or DA-specific manner induces            the SNpc. The α-syn aggregation is then induced by light
α-syn aggregates, mitochondrial fragmentation, motor            stimulation through an optical fiber implanted above the
impairments, and loss of DA neurons. These non-mam-             injection site. Following repeated optogenetic stimula-
malian models offer an interesting way for rapid screen-        tions, the nigrostriatal transmission is compromised,
ing of anti-aggregation targets. However, they might be         leading to Lewy body-like structure formation, neuro-
too simplistic to transpose the results directly to complex     degeneration, and motor impairments within 2 months.
and still not fully understood mechanisms of aggrega-           This inducible model of α-syn aggregation allows for real-
tion, propagation, and toxicity of synucleinopathies in the     time monitoring of α-syn aggregation and Lewy body-like
human brain.                                                    formation with high spatiotemporal control. Altogether,
                                                                viral vectors are very versatile and powerful tools that
Viral vector‑mediated models                                    offer the opportunity to closely mimic PD pathology by
α‑syn viral expression                                          inducing the overexpression of α-syn within multiple
Another alternative to model PD in animals is to overex-        animal species (Table 1). Viral overexpression allows us
press WT or mutated α-syn, including A53T and A30P              to study α-synucleinopathy more precisely and to fol-
forms [297], using lentiviral [298] or AAV vectors [299,        low the progressive development of PD-like phenotypes.
300]. These viral vectors can transduce diverse cell types      Lastly, this tool may also target other non-dopaminergic
and drive the expression of α-syn in the brain region of
Dovonou et al. Translational Neurodegeneration   (2023) 12:36                                                   Page 16 of 25
systems, following the impact of α-syn in the periphery          against PD. However, PD is a highly heterogeneous dis-
or other neuronal cell populations.                              ease involving several factors and pathways that may dif-
                                                                 fer among clinical cases. Therefore, none of the existing
Viral neuromelanin (NM)‑like production                          and future models will replicate the entire spectrum of
Neurodegeneration is particularly prominent within brain         clinical features listed in PD. In this review, we tried to
regions containing NM. In 1919, a visually noticeable loss       summarize the most relevant PD animal models, their
of pigmented neurons in the SNpc was first reported by           respective advantages and disadvantages, as well as their
Konstantin Tretiakoff [320]. This pigmentation loss in the       ability to reproduce the main hallmarks of PD.
brains of PD patients referred to a decrease in NM con-             The choice of the animal model must be based on the
centration, which was less than 50% of the expected levels       PD features addressed by the question the experimenter
for age-matched healthy individuals [321]. NM pigment            seeks to answer. Neurotoxin-induced animal models
appears black and consists of melanin, proteins, lipids,         remain popular due to their cost-effectiveness in generat-
and metal ions. It can be found in different human cen-          ing a PD-like phenotype in a relatively short time span.
tral nervous system neurons and is particularly abundant         Thus, they are commonly used in drug validation for
in DA neurons of the SNpc and noradrenergic neurons of           symptomatic treatment of PD or cell replacement ther-
the locus coeruleus. The pigment itself is contained within      apy. On the other hand, to investigate the function of PD-
specific types of lysosomes fused with autophagic vacu-          linked genes in disease development, transgenic models
oles [322]. Additionally, DA and norepinephrine appear to        should be considered. Other questions concerning the
be the principal substrates in its synthesis. However, NM        targeting of α-syn pathology can also be answered in ani-
may have both a protective and a toxic effect, depending         mal models injected with PFFs or viral vectors encoding
on the conditions of the neuronal environment. Under             SNCA gene copies. In the absence of a suitable model
physiological conditions, NM is proposed to have a pro-          fully recapitulating the targeted PD features of interest,
tective role via iron binding. In PD, iron-bound NM can          a possible solution could be to complement the pheno-
easily release excess iron, producing a toxic effect via         types by combining some of the listed models.
redox processes. Moreover, the iron overload of NM can              The selection of animal species is equally as important.
catalyze DA oxidation, forming DA-o-quinone, and modi-           Although the translational results to humans are valuable
fying proteins that trigger neurotoxic effects [323] (Fig. 2).   in mammalian models, the use of rodents and primates
Yet, the structure and function of human NM in PD                for drug or gene screening is untenable. C. elegans and
have proven to be difficult to investigate due to the lim-       Drosophila could be considered to identify novel neuro-
ited quantities available for extraction from human brain        protective targets [293, 329]. However, the simplicity and
tissues. Furthermore, animal models including rodents            lack of similarity with humans are limitations. Moreo-
lack NM. Nevertheless, recent in vivo models have been           ver, research in PD should not be limited to the use of in
explored, reporting microglial activation via intranigral        vivo models. Although animals offer a better pre-clinical
synthetic NM injections [324–326]. A new approach was            prediction of a drug effect, in vitro models could provide
applied in rodents to virally express human tyrosinase           complementary information on the underlying molecular
(hTyr) in the SNpc. This model shows NM-like pigment             mechanisms [330]. Still, most in vitro-based models can-
accumulation in SNpc DA neurons in an age-dependent              not reproduce the non-cell autonomous impact of the
manner. In fact, overexpression of hTyr recapitulates the        complex cell network in the brain. The near future could
principal hallmarks of PD: mice show mitochondrial and           shed light on the use of human brain organoids to per-
autophagic dysfunction, as well as neuroinflammation.            form drug or gene screenings, although additional char-
They also have impaired DA release, striatal denerva-            acterization of these emerging models will be required
tion, loss of DA neurons in the SNpc, and motor deficits         [330, 331].
by 4 months post-injection [327]. This is accompanied by
Lewy body-like structures and autophagic dysfunction             Conclusion
within NM-like positive neurons [327, 328]. Therefore,           Despite efforts devoted to interpreting the pathogenesis
this model provides a valuable experimental tool to study        of PD, the initial source of neuronal degeneration remains
the effects of NM production and accumulation on neu-            unclear. There is considerable evidence to suggest that
ronal function and viability (Table 1).                          α-syn aggregation, neuroinflammation, as well as lysoso-
                                                                 mal and mitochondrial dysfunction could all play a key
Discussion                                                       role in neurodegeneration [3]. Basic research has yet to
Modelling PD pathogenesis remains a difficult task.              reconcile these features, discriminating causes from con-
Several animal models have been developed to under-              sequences in the PD pathogenesis. Existing animal mod-
stand the pathogenesis and test new drug candidates              els, many of which recapitulate multiple disease features
Dovonou et al. Translational Neurodegeneration            (2023) 12:36                                                                            Page 17 of 25
from cellular pathology and pathogenesis to motor and                             Availability data and materials
                                                                                  Not applicable.
non-motor symptoms, open new possibilities. However,
the wide range available can make choosing a model for
a given study challenging. This task becomes particularly                         Declarations
complex if the study addresses multiple aspects of PD.                            Ethical approval and consent to participate
This review compiles information on the diversity of ani-                         Not applicable.
mal models, and is therefore, a support for researchers to                        Consent for publication
select the optimal animal model for their study. Carefully                        Not applicable.
constructed research models are an essential first step to
                                                                                  Competing interests
understand the disease and discover novel therapies, pos-                         The authors declare that they have no competing interests.
sibly improving the quality of life and the clinical fate of
patients.
                                                                                  Received: 13 March 2023 Accepted: 19 June 2023
Abbreviations
6-OHDA	6-Hydroxydopamine
AAV	Adeno-associated virus
α-syn	α-synuclein
                                                                                  References
BBB	Blood-brain barrier
                                                                                    1. Dorsey ER, Elbaz A, Nichols E, Abd-Allah F, Abdelalim A, Adsuar JC, et al.
DAT	Dopamine transporter
                                                                                        Global, regional, and national burden of Parkinson’s disease, 1990–2016:
DIP	Drug-induced parkinsonism
                                                                                        a systematic analysis for the global burden of Disease Study 2016.
DJ-1	Daisuke-Junko-1 gene
                                                                                        Lancet Neurol. 2018;17:939–53.
dUCH	UCH-L1 ortholog gene
                                                                                    2. Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, et al.
GBA1	β-Glucocerebrosidase
                                                                                        Parkinson disease. Nat Rev Dis Primer. 2017;3:17013.
GCase	Glucocerebrosidase enzyme
                                                                                    3. Kouli A, Torsney KM, Kuan WL. Parkinson’s disease: etiology, neuropa-
hTyr	Human tyrosinase
                                                                                        thology, and pathogenesis. In: Stoker TB, Greenland JC, editors. Par-
KD	Knock-Down
                                                                                        kinson’s disease: pathogenesis and clinical aspects [Internet]. Brisbane
KI	Knock-In
                                                                                        (AU): Codon Publications; 2018.
KO	Knock-Out
                                                                                    4. Postuma RB, Berg D, Stern M, Poewe W, Olanow CW, Oertel W, et al.
L-DOPA	Levodopa or l-3,4-dihydroxyphenylalanine
                                                                                        MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord.
LIPA	light-inducible α-syn aggregation system
                                                                                        2015;30:1591–601.
LPS	Lipopolysaccharide
                                                                                    5. Picca A, Guerra F, Calvani R, Romano R, Coelho-Júnior HJ, Bucci C, et al.
LRRK2	Leucine-rich repeat kinase 2
                                                                                        Mitochondrial dysfunction, protein misfolding and neuroinflammation
MCI	Mitochondrial Complex I
                                                                                        in Parkinson’s disease: roads to biomarker discovery. Biomolecules.
MPP+	1-methyl-4-phenylpyridinium
                                                                                        2021;11:1508.
MPTP	1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
                                                                                    6. Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, et al.
NM	Neuromelanin
                                                                                        In vivo imaging of microglial activation with [11 C](R)-PK11195 PET in
PD	Parkinson’s disease
                                                                                        idiopathic Parkinson’s disease. Neurobiol Dis. 2006;21:404–12.
PDGF-β	Platelet-derived growth factor beta
                                                                                    7. Höllerhage M. Secondary parkinsonism due to drugs, vascular lesions,
PFF	Preformed fibril
                                                                                        tumors, trauma, and other insults. Int Rev Neurobiol. 2019;149:377–418.
PINK1	PTEN-induced putative kinase 1
                                                                                    8. Brigo F, Erro R, Marangi A, Bhatia K, Tinazzi M. Differentiating drug-
POLG	polymerase gamma
                                                                                        induced parkinsonism from Parkinson’s disease: an update on
PRKN	Parkin RBR E3 ubiquitin-protein ligase or PARKIN
                                                                                        non-motor symptoms and investigations. Parkinsonism Relat Disord.
Prp	Prion protein
                                                                                        2014;20:808–14.
ROS	Reactive oxygen species
                                                                                    9. López-Sendón J, Mena MA, de Yébenes JG. Drug-induced parkinsonism.
SNpc	Substantia nigra pars compacta
                                                                                        Expert Opin Drug Saf. 2013;12:487–96.
Tau	tubulin-associated unit
                                                                                   10. Carlsson A, Lindqvist M, Magnusson T. 3,4-Dihydroxyphenylala-
TH	Tyrosine hydroxylase
                                                                                        nine and 5-hydroxytryptophan as reserpine antagonists. Nature.
Thy1	Thymocyte differentiation antigen 1
                                                                                        1957;180:1200.
UCH-L1	Ubiquitin carboxy-terminal hydrolase L1
                                                                                   11. Bernstein AI, Stout KA, Miller GW. The vesicular monoamine trans-
VTA	Ventral tegmental area
                                                                                        porter 2: an underexplored pharmacological target. Neurochem Int.
                                                                                        2014;73:89–97.
                                                                                   12. Freis ED. Mental depression in hypertensive patients treated for long
Acknowledgements
                                                                                        periods with large doses of reserpine. N Engl J Med. 1954;251:1006–8.
Not applicable.
                                                                                   13. Freis ED, Ari R. Clinical and experimental effects of reserpine in patients
                                                                                        with essential hypertension. Ann N Y Acad Sci. 1954;59:45–53.
Author contributions
                                                                                   14. Kane JM, Smith JM. Tardive dyskinesia: prevalence and risk factors, 1959
AD: writing, review and editing. CB: writing, review and editing. VSL: writing,
                                                                                        to 1979. Arch Gen Psychiatry. 1982;39:473–81.
review and editing. CG: writing, review and editing. MRP 3rd: review and edit-
                                                                                   15. Fernandes VS, Santos JR, Leão AHFF, Medeiros AM, Melo TG, Izídio GS,
ing. ML: funding acquisition, writing, review and editing. All authors read and
                                                                                        et al. Repeated treatment with a low dose of reserpine as a progressive
approved the final manuscript.
                                                                                        model of Parkinson’s disease. Behav Brain Res. 2012;231:154–63.
                                                                                   16. Santos JR, Cunha JAS, Dierschnabel AL, Campêlo CLC, Leão AHFF, Silva
Funding
                                                                                        AF, et al. Cognitive, motor and tyrosine hydroxylase temporal impair-
This work was supported by a grant to ML. from Canadian Institutes of Health
                                                                                        ment in a model of parkinsonism induced by reserpine. Behav Brain
Research. ML. also receives a salary award from the Fonds de recherche du
                                                                                        Res. 2013;253:68–77.
Québec in partnership with Parkinson Quebec.
Dovonou et al. Translational Neurodegeneration            (2023) 12:36                                                                             Page 18 of 25
 17. Bispo JMM, Melo JEC, Gois AM, Leal PC, Lins LCRF, Souza MF, et al. Sex        40. Blum D, Torch S, Lambeng N, Nissou M, Benabid AL, Sadoul R, et al.
     differences in the progressive model of parkinsonism induced by                   Molecular pathways involved in the neurotoxicity of 6-OHDA, dopa-
     reserpine in rats. Behav Brain Res. 2019;363:23–9.                                mine and MPTP: contribution to the apoptotic theory in Parkinson’s
 18. Leão AHFF, Meurer YSR, da Silva AF, Medeiros AM, Campêlo CLC, Abílio              disease. Prog Neurobiol. 2001;65:135–72.
     VC, et al. Spontaneously hypertensive rats (SHR) are resistant to a           41. Schober A. Classic toxin-induced animal models of Parkinson’s disease:
     reserpine-induced progressive model of Parkinson’s disease: differences           6-OHDA and MPTP. Cell Tissue Res. 2004;318:215–24.
     in motor behavior, tyrosine hydroxylase and α-synuclein expression.           42. Walsh S, Finn DP, Dowd E. Time-course of nigrostriatal neurodegen-
     Front Aging Neurosci. 2017;9:78.                                                  eration and neuroinflammation in the 6-hydroxydopamine-induced
 19. Leão AHFF, Sarmento-Silva AJ, Santos JR, Ribeiro AM, Silva RH.                    axonal and terminal lesion models of Parkinson’s disease in the rat.
     Molecular, neurochemical, and behavioral hallmarks of reserpine as                Neuroscience. 2011;175:251–61.
     a model for Parkinson’s disease: new perspectives to a long-standing          43. Hernandez-Baltazar D, Zavala-Flores LM, Villanueva-Olivo A. The
     model: reserpine as a model of Parkinson’s disease. Brain Pathol.                 6-hydroxydopamine model and parkinsonian pathophysiology: novel
     2015;25:377–90.                                                                   findings in an older model. Neurol Barc Spain. 2017;32:533–9.
 20. Lama J, Buhidma Y, Fletcher EJR, Duty S. Animal models of Parkinson’s         44. Kirik D, Rosenblad C, Björklund A. Characterization of behavioral and
     disease: a guide to selecting the optimal model for your research.                neurodegenerative changes following partial lesions of the nigrostriatal
     Neuronal Signal. 2021;5:NS20210026.                                               dopamine system induced by intrastriatal 6-hydroxydopamine in the
 21. Leal PC, Lins LCRF, de Gois AM, Marchioro M, Santos JR. Commen-                   rat. Exp Neurol. 1998;152:259–77.
     tary: evaluation of models of Parkinson’s disease. Front Neurosci.            45. Deumens R, Blokland A, Prickaerts J. Modeling Parkinson’s disease in
     2016;10:283.                                                                      rats: an evaluation of 6-OHDA lesions of the nigrostriatal pathway. Exp
 22. Gardner DM. Modern antipsychotic drugs: a critical overview. Can Med              Neurol. 2002;175:303–17.
     Assoc J. 2005;172:1703–11.                                                    46. Stott SRW, Barker RA. Time course of dopamine neuron loss and glial
 23. Ezrin-Waters C, Muller P, Seeman P. Catalepsy induced by morphine or              response in the 6-OHDA striatal mouse model of Parkinson’s disease.
     haloperidol: effects of apomorphine and anticholinergic drugs. Can J              Eur J Neurosci. 2014;39:1042–56.
     Physiol Pharmacol. 1976;54:516–9.                                             47. Sauer H, Oertel WH. Progressive degeneration of nigrostriatal dopamine
 24. Sanberg PR. Haloperidol-induced catalepsy is mediated by postsynap-               neurons following intrastriatal terminal lesions with 6-hydroxydopa-
     tic dopamine receptors. Nature. 1980;284:472–3.                                   mine: a combined retrograde tracing and immunocytochemical study
 25. Kulkarni SK, Bishnoi M, Chopra K. In vivo microdialysis studies of striatal       in the rat. Neuroscience. 1994;59:401–15.
     level of neurotransmitters after haloperidol and chlorpromazine admin-        48. Branchi I, D’Andrea I, Armida M, Cassano T, Pèzzola A, Potenza RL, et al.
     istration. Indian J Exp Biol. 2009;47:91–7.                                       Nonmotor symptoms in Parkinson’s disease: investigating early-phase
 26. Balijepalli S, Kenchappa RS, Boyd MR, Ravindranath V. Protein thiol               onset of behavioral dysfunction in the 6-hydroxydopamine-lesioned rat
     oxidation by haloperidol results in inhibition of mitochondrial complex           model. J Neurosci Res. 2008;86:2050–61.
     I in brain regions: comparison with atypical antipsychotics. Neurochem        49. Colucci M, Cervio M, Faniglione M, De Angelis S, Pajoro M, Levandis G,
     Int. 2001;38:425–35.                                                              et al. Intestinal dysmotility and enteric neurochemical changes in a Par-
 27. Ionov ID, Severtsev NN. Somatostatin antagonist potentiates halop-                kinson’s disease rat model. Auton Neurosci Basic Clin. 2012;169:77–86.
     eridol-induced catalepsy in the aged rat. Pharmacol Biochem Behav.            50. Pellegrini C, Ippolito C, Segnani C, Dolfi A, Errede M, Virgintino D, et al.
     2012;103:295–8.                                                                   Pathological remodelling of colonic wall following dopaminergic
 28. Ionov ID, Severtsev NN. Histamine- and haloperidol-induced catalepsy              nigrostriatal neurodegeneration. Neurobiol Dis. 2020;139:104821.
     in aged mice: differential responsiveness to L-DOPA. Psychopharmacol-         51. Domenici RA, Campos ACP, Maciel ST, Berzuino MB, Hernandes MS,
     ogy. 2012;223:191–7.                                                              Fonoff ET, et al. Parkinson’s disease and pain: modulation of noci-
 29. Sharma AK, Gupta S, Patel RK, Wardhan N. Haloperidol-induced                      ceptive circuitry in a rat model of nigrostriatal lesion. Exp Neurol.
     parkinsonism is attenuated by varenicline in mice. J Basic Clin Physiol           2019;315:72–81.
     Pharmacol. 2018;29:395–401.                                                   52. He X, Yuan W, Li Z, Hou Y, Liu F, Feng J. 6-Hydroxydopamine induces
 30. Waku I, Magalhães MS, Alves CO, de Oliveira AR. Haloperidol-induced               autophagic flux dysfunction by impairing transcription factor EB activa-
     catalepsy as an animal model for parkinsonism: a systematic review of             tion and lysosomal function in dopaminergic neurons and SH-SY5Y
     experimental studies. Eur J Neurosci. 2021;53:3743–67.                            cells. Toxicol Lett. 2018;283:58–68.
 31. Blanchet PJ, Parent M-T, Rompré PH, Lévesque D. Relevance of animal           53. Nass R, Hall DH, Miller DM, Blakely RD. Neurotoxin-induced degenera-
     models to human tardive dyskinesia. Behav Brain Funct BBF. 2012;8:12.             tion of dopamine neurons in Caenorhabditis elegans. Proc Natl Acad
 32. Gunne LM, Bárány S. Haloperidol-induced tardive dyskinesia in mon-                Sci U S A. 2002;99:3264–9.
     keys. Psychopharmacology. 1976;50:237–40.                                     54. Hernandez-Baltazar D, Mendoza-Garrido ME, Martinez-Fong D. Activa-
 33. Mahmoudi S, Blanchet PJ, Lévesque D. Haloperidol-induced striatal                 tion of GSK-3β and caspase-3 occurs in Nigral dopamine neurons
     Nur77 expression in a non-human primate model of tardive dyskinesia.              during the development of apoptosis activated by a striatal injection of
     Eur J Neurosci. 2013;38:2192–8.                                                   6-hydroxydopamine. PLoS ONE. 2013;8:e70951.
 34. Varty GB, Hodgson RA, Pond AJ, Grzelak ME, Parker EM, Hunter JC.              55. Lee Mosley R, Benner EJ, Kadiu I, Thomas M, Boska MD, Hasan K, et al.
     The effects of adenosine A2A receptor antagonists on haloperidol-                 Neuroinflammation, oxidative stress, and the pathogenesis of Parkin-
     induced movement disorders in primates. Psychopharmacology.                       son’s disease. Clin Neurosci Res. 2006;6:261–81.
     2008;200:393–401.                                                             56. Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic parkinsonism in
 35. Thakur KS, Prakash A, Bisht R, Bansal PK. Beneficial effect of candesartan        humans due to a product of meperidine-analog synthesis. Science.
     and lisinopril against haloperidol-induced tardive dyskinesia in rat. J           1983;219:979–80.
     Renin-Angiotensin-Aldosterone Syst JRAAS. 2015;16:917–29.                     57. Nicklas WJ, Youngster SK, Kindt MV, Heikkila RE. IV. MPTP, MPP + and
 36. Senoh S, Witkop B. Non-enzymatic conversions of dopamine to                       mitochondrial function. Life Sci. 1987;40:721–9.
     norepinephrine and trihydroxyphenethylamines. J Am Chem Soc.                  58. Przedborski S, Vila M. The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
     1959;81:6222–31.                                                                  mouse model: a tool to explore the pathogenesis of Parkinson’s disease.
 37. Blandini F, Armentero M-T, Martignoni E. The 6-hydroxydopamine                    Ann N Y Acad Sci. 2006;991:189–98.
     model: news from the past. Parkinsonism Relat Disord. 2008;14(Suppl           59. Lofrumento DD, Saponaro C, Cianciulli A, De Nuccio F, Mitolo V, Nico-
     2):124–9.                                                                         lardi G, et al. MPTP-induced neuroinflammation increases the expres-
 38. Hernandez-Baltazar D, Zavala-Flores LM, Villanueva-Olivo A. The                   sion of pro-inflammatory cytokines and their receptors in mouse brain.
     6-hydroxydopamine model and parkinsonian pathophysiology: novel                   Neuroimmunomodulation. 2011;18:79–88.
     findings in an older model. Neurol Engl Ed Elsevier. 2017;32:533–9.           60. Przedborski S, Jackson-Lewis V, Naini AB, Jakowec
 39. Ungerstedt U. 6-hydroxy-dopamine induced degeneration of central                  M, Petzinger G, Miller R, et al. The parkinsonian toxin
     monoamine neurons. Eur J Pharmacol. 1968;5:107–10.
Dovonou et al. Translational Neurodegeneration           (2023) 12:36                                                                               Page 19 of 25
       1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): a technical review     83. Wang J, He C, Wu WY, Chen F, Wu YY, Li WZ, et al. Biochanin A protects
       of its utility and safety: the parkinsonian toxin MPTP. J Neurochem.            dopaminergic neurons against lipopolysaccharide-induced damage
       2001;76:1265–74.                                                                and oxidative stress in a rat model of Parkinson’s disease. Pharmacol
 61.   Ding F, Luan L, Ai Y, Walton A, Gerhardt GA, Gash DM, et al. Develop-           Biochem Behav. 2015;138:96–103.
       ment of a stable, early stage unilateral model of Parkinson’s disease in    84. Zhang J, Stanton DM, Nguyen XV, Liu M, Zhang Z, Gash D, et al. Intrapal-
       middle-aged rhesus monkeys. Exp Neurol. 2008;212:431–9.                         lidal lipopolysaccharide injection increases iron and ferritin levels in glia
 62.   Schmidt N, Ferger B. Neurochemical findings in the MPTP model of                of the rat substantia nigra and induces locomotor deficits. Neurosci-
       Parkinson’s disease. J Neural Transm. 2001;108:1263–82.                         ence. 2005;135:829–38.
 63.   Hu S, Hu M, Liu J, Zhang B, Zhang Z, Zhou FH, et al. Phosphorylation        85. Zhang FX, Xu RS. Juglanin ameliorates LPS-induced neuroinflammation
       of tau and α-synuclein induced neurodegeneration in MPTP mouse                  in animal models of Parkinson’s disease and cell culture via inactivating
       model of Parkinson’s disease. Neuropsychiatr Dis Treat. 2020;16:651–63.         TLR4/NF-κB pathway. Biomed Pharmacother. 2018;97:1011–9.
 64.   Gelders G, Baekelandt V, Van der Perren A. Linking neuroinflamma-           86. Choi DY, Zhang J, Bing G. Aging enhances the neuroinflammatory
       tion and neurodegeneration in Parkinson’s disease. J Immunol Res.               response and alpha-synuclein nitration in rats. Neurobiol Aging.
       2018;2018:1–12.                                                                 2010;31:1649–53.
 65.   Liu M, Bing G. Lipopolysaccharide animal models for Parkinson’s dis-        87. Liu Y, Qin L, Wilson B, Wu X, Qian L, Granholm AC, et al. Endotoxin
       ease. Park Dis. 2011;2011:327089.                                               induces a delayed loss of TH-IR neurons in substantia nigra and motor
 66.   Bertani B, Ruiz N. Function and biogenesis of lipopolysaccharides.              behavioral deficits. Neurotoxicology. 2008;29:864–70.
       EcoSal Plus. 2018;8.                                                        88. Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, et al. Systemic LPS
 67.   Seemann S, Zohles F, Lupp A. Comprehensive comparison of three                  causes chronic neuroinflammation and progressive neurodegenera-
       different animal models for systemic inflammation. J Biomed Sci.                tion. Glia. 2007;55:453–62.
       2017;24:60.                                                                 89. Zheng HF, Yang YP, Hu LF, Wang MX, Wang F, Cao LD, et al. Autophagic
 68.   Niehaus I. Endotoxin: is it an environmental factor in the cause of             impairment contributes to systemic inflammation-induced dopaminer-
       Parkinson’s disease? Occup Environ Med. 2003;60:378–8.                          gic neuron loss in the midbrain. PLoS ONE. 2013;8:e70472.
 69.   Castaño A, Herrera AJ, Cano J, Machado A. Lipopolysaccharide                90. Li YH, He Q, Yu J, Liu C, Feng L, Chai Z, et al. Lipoic acid protects dopa-
       intranigral injection induces inflammatory reaction and damage in               minergic neurons in LPS-induced Parkinson’s disease model. Metab
       nigrostriatal dopaminergic system. J Neurochem. 1998;70:1584–92.                Brain Dis. 2015;30:1217–26.
 70.   Bing GY, Lu X, Zheng NY, Jin L, Floyd RA, Kim HC. Microglia mediated        91. Zhao YF, Qiong-Zhang, Zhang JF, Lou ZY, Zu HB, Wang ZG, et al. The
       dopaminergic cell death in the substantia nigra: a new animal model             synergy of aging and LPS exposure in a mouse model of Parkinson’s
       for Parkinson’s disease. Free Radic Biol Med. 1998;25:S44–4.                    disease. Aging Dis. 2018;9:785.
 71.   Herrera AJ, Castaño A, Venero JL, Cano J, Machado A. The single intrani-    92. Nazem A, Sankowski R, Bacher M, Al-Abed Y. Rodent models of neuroin-
       gral injection of LPS as a new model for studying the selective effects         flammation for Alzheimer’s disease. J Neuroinflammation. 2015;12:74.
       of inflammatory reactions on dopaminergic system. Neurobiol Dis.            93. Roe AD, Staup MA, Serrats J, Sawchenko PE, Rissman RA. Lipopolysac-
       2000;7:429–47.                                                                  charide-induced tau phosphorylation and kinase activity - modulation,
 72.   Deng I, Corrigan F, Zhai G, Zhou X-F, Bobrovskaya L. Lipopolysaccharide         but not mediation, by corticotropin-releasing factor receptors: LPS-
       animal models of Parkinson’s disease: recent progress and relevance to          induced tau-P. Eur J Neurosci. 2011;34:448–56.
       clinical disease. Brain Behav Immun. 2020;4:100060.                         94. Valero J, Mastrella G, Neiva I, Sánchez S, Malva JO. Long-term effects of
 73.   Hunter RL, Cheng B, Choi DY, Liu M, Liu S, Cass WA, et al. Intrastriatal        an acute and systemic administration of LPS on adult neurogenesis and
       lipopolysaccharide injection induces parkinsonism in C57/B6 mice. J             spatial memory. Front Neurosci. 2014;8.
       Neurosci Res. 2009;87:1913–21.                                              95. Wang LM, Wu Q, Kirk RA, Horn KP, Ebada Salem AH, Hoffman JM, et al.
 74.   Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, Lee VM-Y.               Lipopolysaccharide endotoxemia induces amyloid-β and p-tau forma-
       Neuroinflammation and oxidation/nitration of alpha-synuclein linked             tion in the rat brain. Am J Nucl Med Mol Imaging. 2018;8:86–99.
       to dopaminergic neurodegeneration. J Neurosci. 2008;28:7687–98.             96. Ascherio A, Schwarzschild MA. The epidemiology of Parkinson’s disease:
 75.   Qin L, Liu Y, Wang T, Wei S-J, Block ML, Wilson B, et al. NADPH oxidase         risk factors and prevention. Lancet Neurol. 2016;15:1257–72.
       mediates lipopolysaccharide-induced neurotoxicity and proin-                97. Baldi I, Cantagrel A, Lebailly P, Tison F, Dubroca B, Chrysostome V, et al.
       flammatory gene expression in activated microglia. J Biol Chem.                 Association between Parkinson’s disease and exposure to pesticides in
       2004;279:1415–21.                                                               southwestern France. Neuroepidemiology. 2003;22:305–10.
 76.   Bao LH, Zhang YN, Zhang JN, Gu L, Yang HM, Huang YY, et al. Urate           98. Petrovitch H, Ross GW, Abbott RD, Sanderson WT, Sharp DS, Tanner
       inhibits microglia activation to protect neurons in an LPS-induced              CM, et al. Plantation work and risk of Parkinson disease in a population-
       model of Parkinson’s disease. J Neuroinflammation. 2018;15:131.                 based longitudinal study. Arch Neurol. 2002;59:1787–92.
 77.   Chen G, Liu J, Jiang L, Ran X, He D, Li Y, et al. Galangin reduces the      99. Costello S, Cockburn M, Bronstein J, Zhang X, Ritz B. Parkinson’s disease
       loss of dopaminergic neurons in an LPS-evoked model of Parkinson’s              and residential exposure to maneb and paraquat from agricultural
       disease in rats. Int J Mol Sci. 2017;19:12.                                     applications in the central valley of California. Am J Epidemiol.
 78.   Choi DY, Liu M, Hunter RL, Cass WA, Pandya JD, Sullivan PG, et al.              2009;169:919–26.
       Striatal neuroinflammation promotes parkinsonism in rats. PLoS ONE.        100. Tanner CM, Kamel F, Ross GW, Hoppin JA, Goldman SM, Korell M, et al.
       2009;4:e5482.                                                                   Rotenone, paraquat, and Parkinson’s disease. Environ Health Perspect.
 79.   Fu SP, Wang JF, Xue WJ, Liu HM, Liu B, Zeng YL, et al. Anti-inflammatory        2011;119:866–72.
       effects of BHBA in both in vivo and in vitro Parkinson’s disease models    101. Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greena-
       are mediated by GPR109A-dependent mechanisms. J Neuroinflamma-                  myre JT. Chronic systemic pesticide exposure reproduces features of
       tion. 2015;12:9.                                                                Parkinson’s disease. Nat Neurosci. 2000;3:1301–6.
 80.   He H, Guo WW, Xu RR, Chen XQ, Zhang N, Wu X, et al. Alkaloids from         102. Mader BJ, Pivtoraiko VN, Flippo HM, Klocke BJ, Roth KA, Mangieri LR,
       piper longum protect dopaminergic neurons against inflammation-                 et al. Rotenone inhibits autophagic flux prior to inducing cell death.
       mediated damage induced by intranigral injection of lipopolysaccha-             ACS Chem Neurosci. 2012;3:1063–72.
       ride. BMC Complement Altern Med. 2016;16:412.                              103. Yuan YH, Yan WF, Sun JD, Huang JY, Mu Z, Chen NH. The molecu-
 81.   Hunter R, Ojha U, Bhurtel S, Bing G, Choi DY. Lipopolysaccharide-               lar mechanism of rotenone-induced α-synuclein aggregation:
       induced functional and structural injury of the mitochondria in the             emphasizing the role of the calcium/GSK3β pathway. Toxicol Lett.
       nigrostriatal pathway. Neurosci Res. 2017;114:62–9.                             2015;233:163–71.
 82.   Hunter RL, Dragicevic N, Seifert K, Choi DY, Liu M, Kim HC, et al.         104. Fleming S. Behavioral and immunohistochemical effects of chronic
       Inflammation induces mitochondrial dysfunction and dopaminer-                   intravenous and subcutaneous infusions of varying doses of rotenone.
       gic neurodegeneration in the nigrostriatal system. J Neurochem.                 Exp Neurol. 2004;187:418–29.
       2007;100:1375–86.                                                          105. Inden M, Kitamura Y, Takeuchi H, Yanagida T, Takata K, Kobayashi
                                                                                       Y, et al. Neurodegeneration of mouse nigrostriatal dopaminergic
Dovonou et al. Translational Neurodegeneration            (2023) 12:36                                                                             Page 20 of 25
       system induced by repeated oral administration of rotenone is               127. Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015;386:896–912.
       prevented by 4-phenylbutyrate, a chemical chaperone. J Neurochem.           128. Blauwendraat C, Nalls MA, Singleton AB. The genetic architecture of
       2007;101:1491–504.                                                               Parkinson’s disease. Lancet Neurol Elsevier. 2020;19:170–8.
106.   Whitton PS. Inflammation as a causative factor in the aetiology of          129. Pickrell AM, Youle RJ. The roles of PINK1, parkin, and mitochondrial fidel-
       Parkinson’s disease. Br J Pharmacol. 2007;150:963–76.                            ity in Parkinson’s disease. Neuron. 2015;85:257–73.
107.   Tansey MG, Goldberg MS. Neuroinflammation in Parkinson’s disease: its       130. Schneider SA, Alcalay RN. Neuropathology of genetic synucle-
       role in neuronal death and implications for therapeutic intervention.            inopathies with parkinsonism: review of the literature. Mov Disord.
       Neurobiol Dis. 2010;37:510–8.                                                    2017;32:1504–23.
108.   Xiong N, Xiong J, Jia M, Liu L, Zhang X, Chen Z, et al. The role of         131. Cooper JF, Machiela E, Dues DJ, Spielbauer KK, Senchuk MM, Van
       autophagy in Parkinson’s disease: rotenone-based modeling. Behav                 Raamsdonk JM. Activation of the mitochondrial unfolded protein
       Brain Funct. 2013;9:13.                                                          response promotes longevity and dopamine neuron survival in Parkin-
109.   Johnson ME, Bobrovskaya L. An update on the rotenone models of                   son’s disease models. Sci Rep. 2017;7:16441.
       Parkinson’s disease: their ability to reproduce the features of clinical    132. Cooper JF, Van Raamsdonk JM. Modeling Parkinson’s disease in C.
       disease and model gene-environment interactions. Neurotoxicology.                elegans. J Park Dis. 2018;8:17–32.
       2015;46:101–16.                                                             133. Park J, Lee SB, Lee S, Kim Y, Song S, Kim S, et al. Mitochondrial dysfunc-
110.   Cicchetti F, Drouin-Ouellet J, Gross RE. Viability of the rotenone model         tion in Drosophila PINK1 mutants is complemented by parkin. Nature.
       in question. Trends Pharmacol Sci. 2010;31:142–3.                                2006;441:1157–61.
111.   Greenamyre JT, Cannon JR, Drolet R, Mastroberardino PG. Lessons             134. Greene JC, Whitworth AJ, Kuo I, Andrews LA, Feany MB, Pallanck LJ.
       from the rotenone model of Parkinson’s disease. Trends Pharmacol Sci.            Mitochondrial pathology and apoptotic muscle degeneration in Dros-
       2010;31:141–2. author reply 142–143.                                             ophila parkin mutants. Proc Natl Acad Sci U S A. 2003;100:4078–83.
112.   Soderstrom K, O’Malley J, Steece-Collier K, Kordower JH. Neural repair      135. Ge P, Dawson VL, Dawson TM. PINK1 and parkin mitochondrial quality
       strategies for Parkinson’s disease: insights from primate models. Cell           control: a source of regional vulnerability in Parkinson’s disease. Mol
       Transpl. 2006;15:251–65.                                                         Neurodegener. 2020;15:20.
113.   Berry C, La Vecchia C, Nicotera P. Paraquat and Parkinson’s disease. Cell   136. Dawson TM, Ko HS, Dawson VL. Genetic animal models of Parkinson’s
       Death Differ. 2010;17:1115–25.                                                   disease. Neuron. 2010;66:646–61.
114.   Rappold PM, Cui M, Chesser AS, Tibbett J, Grima JC, Duan L,                 137. Noda S, Sato S, Fukuda T, Tada N, Uchiyama Y, Tanaka K, et al. Loss of par-
       et al. Paraquat neurotoxicity is mediated by the dopamine trans-                 kin contributes to mitochondrial turnover and dopaminergic neuronal
       porter and organic cation transporter-3. Proc Natl Acad Sci U S A.               loss in aged mice. Neurobiol Dis. 2020;136:104717.
       2011;108:20766–71.                                                          138. Yang W, Li S, Li X-J. A CRISPR monkey model unravels a unique function
115.   Cicchetti F, Lapointe N, Roberge-Tremblay A, Saint-Pierre M, Jimenez L,          of PINK1 in primate brains. Mol Neurodegener. 2019;14:17.
       Ficke BW, et al. Systemic exposure to paraquat and maneb models early       139. Xiong Y, Dawson TM, Dawson VL. Models of LRRK2-associated Parkin-
       Parkinson’s disease in young adult rats. Neurobiol Dis. 2005;20:360–71.          son’s disease. Adv Neurobiol. 2017;14:163–91.
116.   McCormack AL, Thiruchelvam M, Manning-Bog AB, Thiffault C, Langston         140. Wallings R, Manzoni C, Bandopadhyay R. Cellular processes associated
       JW, Cory-Slechta DA, et al. Environmental risk factors and Parkinson’s           with LRRK2 function and dysfunction. FEBS J. 2015;282:2806–26.
       disease: selective degeneration of nigral dopaminergic neurons caused       141. Di Maio R, Hoffman EK, Rocha EM, Keeney MT, Sanders LH, De Miranda
       by the herbicide paraquat. Neurobiol Dis. 2002;10:119–27.                        BR, et al. LRRK2 activation in idiopathic Parkinson’s disease. Sci Transl
117.   Thiruchelvam M, Brockel BJ, Richfield EK, Baggs RB, Cory-Slechta DA.             Med. 2018;10:eaar5429.
       Potentiated and preferential effects of combined paraquat and maneb         142. Kalia LV, Lang AE, Hazrati L-N, Fujioka S, Wszolek ZK, Dickson DW, et al.
       on nigrostriatal dopamine systems: environmental risk factors for                Clinical correlations with Lewy body pathology in LRRK2-related Parkin-
       Parkinson’s disease? Brain Res. 2000;873:225–34.                                 son disease. JAMA Neurol. 2015;72:100–5.
118.   Thiruchelvam M, McCormack A, Richfield EK, Baggs RB, Tank AW, Di            143. Poulopoulos M, Levy OA, Alcalay RN. The neuropathology of genetic
       Monte DA, et al. Age-related irreversible progressive nigrostriatal              Parkinson’s disease. Mov Disord. 2012;27:831–42.
       dopaminergic neurotoxicity in the paraquat and maneb model of the           144. Saha S, Guillily MD, Ferree A, Lanceta J, Chan D, Ghosh J, et al. LRRK2
       Parkinson’s disease phenotype. Eur J Neurosci. 2003;18:589–600.                  modulates vulnerability to mitochondrial dysfunction in Caenorhabditis
119.   Manning-Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Di Monte                elegans. J Neurosci. 2009;29:9210–8.
       DA. The herbicide paraquat causes up-regulation and aggregation of          145. Yao C, El Khoury R, Wang W, Byrd TA, Pehek EA, Thacker C, et al. LRRK2-
       alpha-synuclein in mice: paraquat and alpha-synuclein. J Biol Chem.              mediated neurodegeneration and dysfunction of dopaminergic neu-
       2002;277:1641–4.                                                                 rons in a Caenorhabditis elegans model of Parkinson’s disease. Neurobiol
120.   Manning-Bog AB, McCormack AL, Purisai MG, Bolin LM. Di Monte DA.                 Dis. 2010;40:73–81.
       α-Synuclein overexpression protects against paraquat-induced neuro-         146. Saha S, Ash PEA, Gowda V, Liu L, Shirihai O, Wolozin B. Mutations in
       degeneration. J Neurosci. 2003;23:3095–9.                                        LRRK2 potentiate age-related impairment of autophagic flux. Mol
121.   Mitra S, Chakrabarti N, Bhattacharyya A. Differential regional expres-           Neurodegener. 2015;10:26.
       sion patterns of α-synuclein, TNF-α, and IL-1β; and variable status of      147. Angeles DC, Ho P, Chua LL, Wang C, Yap YW, Ng C, et al. Thiol
       dopaminergic neurotoxicity in mouse brain after paraquat treatment. J            peroxidases ameliorate LRRK2 mutant-induced mitochondrial and
       Neuroinflammation. 2011;8:163.                                                   dopaminergic neuronal degeneration in Drosophila. Hum Mol Genet.
122.   Hemmati AA, Hicks R. Increased myofibroblast contractile sensitivity in          2014;23:3157–65.
       paraquat pretreated rat lung tissue. Life Sci. 1999;65:2325–32.             148. Bardai FH, Ordonez DG, Bailey RM, Hamm M, Lewis J, Feany MB. Lrrk
123.   Saint-Pierre M, Tremblay M-E, Sik A, Gross RE, Cicchetti F. Temporal             promotes tau neurotoxicity through dysregulation of actin and mito-
       effects of paraquat/maneb on microglial activation and dopamine                  chondrial dynamics. PLOS Biol. 2018;16:e2006265.
       neuronal loss in older rats. J Neurochem. 2006;98:760–72.                   149. Dodson MW, Zhang T, Jiang C, Chen S, Guo M. Roles of the Drosophila
124.   Thiruchelvam M, Richfield EK, Baggs RB, Tank AW, Cory-Slechta DA. The            LRRK2 homolog in Rab7-dependent lysosomal positioning. Hum Mol
       nigrostriatal dopaminergic system as a preferential target of repeated           Genet. 2012;21:1350–63.
       exposures to combined paraquat and maneb: implications for Parkin-          150. Beccano-Kelly DA, Kuhlmann N, Tatarnikov I, Volta M, Munsie LN, Chou
       son’s disease. J Neurosci Off J Soc Neurosci. 2000;20:9207–14.                   P et al. Synaptic function is modulated by LRRK2 and glutamate release
125.   Soares JJ, Rodrigues DT, Gonçalves MB, Lemos MC, Gallarreta MS,                  is increased in cortical neurons of G2019S LRRK2 knock-in mice. Front
       Bianchini MC, et al. Paraquat exposure-induced Parkinson’s disease-like          Cell Neurosci. 2014;8.
       symptoms and oxidative stress in Drosophila melanogaster: neuropro-         151. Beccano-Kelly DA, Volta M, Munsie LN, Paschall SA, Tatarnikov I, Co K,
       tective effect of Bougainvillea glabra Choisy. Biomed Pharmacother               et al. LRRK2 overexpression alters glutamatergic presynaptic plasticity,
       Biomedecine Pharmacother. 2017;95:245–51.
126.   Cooper JF, Van Raamsdonk JM. Modeling Parkinson’s disease in C.
       elegans. J Park Dis. 2018;8:17–32.
Dovonou et al. Translational Neurodegeneration              (2023) 12:36                                                                             Page 21 of 25
       striatal dopamine tone, postsynaptic signal transduction, motor activity       173. Leroy E, Boyer R, Auburger G, Leube B, Ulm G, Mezey E, et al. The ubiqui-
       and memory. Hum Mol Genet. 2015;24:1336–49.                                         tin pathway in Parkinson’s disease. Nature. 1998;395:451–2.
152.   Li X, Patel JC, Wang J, Avshalumov MV, Nicholson C, Buxbaum JD, et al.         174. Lowe J, McDermott H, Landon M, Mayer RJ, Wilkinson KD. Ubiquitin
       Enhanced striatal dopamine transmission and motor performance                       carboxyl-terminal hydrolase (PGP 9.5) is selectively present in ubiqui-
       with LRRK2 overexpression in mice is eliminated by familial Parkinson’s             tinated inclusion bodies characteristic of human neurodegenerative
       disease mutation G2019S. J Neurosci. 2010;30:1788–97.                               diseases. J Pathol. 1990;161:153–60.
153.   Li Y, Liu W, Oo TF, Wang L, Tang Y, Jackson-Lewis V, et al. Mutant             175. Yasuda T, Nihira T, Ren Y-R, Cao X-Q, Wada K, Setsuie R, et al. Effects of
       LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of                 UCH-L1 on α-synuclein over-expression mouse model of Parkinson’s
       Parkinson’s disease. Nat Neurosci. 2009;12:826–8.                                   disease. J Neurochem. 2009;108:932–44.
154.   Melrose HL, Dächsel JC, Behrouz B, Lincoln SJ, Yue M, Hinkle KM, et al.        176. Cerqueira FM, von Stockum S, Giacomello M, Goliand I, Kakimoto P,
       Impaired dopaminergic neurotransmission and microtubule-associated                  Marchesan E, et al. A new target for an old DUB: UCH-L1 regulates mito-
       protein tau alterations in human LRRK2 transgenic mice. Neurobiol Dis.              fusin-2 levels, altering mitochondrial morphology, function and calcium
       2010;40:503–17.                                                                     uptake. Redox Biol. 2020;37:101676.
155.   Volta M, Beccano-Kelly DA, Paschall SA, Cataldi S, MacIsaac SE, Kuhl-          177. Miura H, Oda K, Endo C, Yamazaki K, Shibasaki H, Kikuchi T. Progres-
       mann N, et al. Initial elevations in glutamate and dopamine neuro-                  sive degeneration of motor nerve terminals in GAD mutant mouse
       transmission decline with age, as does exploratory behavior, in LRRK2               with hereditary sensory axonopathy. Neuropathol Appl Neurobiol.
       G2019S knock-in mice. eLife. 2017;6:e28377.                                         1993;19:41–51.
156.   Yue M, Hinkle KM, Davies P, Trushina E, Fiesel FC, Christenson TA, et al.      178. Oda K, Yamazakit K, Miura H, Shibasaki H, Kikuchi T. Dying back type
       Progressive dopaminergic alterations and mitochondrial abnormalities                axonal degeneration of sensory nerve terminals in muscle spindles of
       in LRRK2 G2019S knock-in mice. Neurobiol Dis. 2015;78:172–95.                       the gracile axonal dystrophy (GAD) mutant mouse. Neuropathol Appl
157.   Chen CY, Weng YH, Chien KY, Lin KJ, Yeh TH, Cheng YP, et al. (G2019S)               Neurobiol. 1992;18:265–81.
       LRRK2 activates MKK4-JNK pathway and causes degeneration of SN                 179. Saigoh K, Wang YL, Suh JG, Yamanishi T, Sakai Y, Kiyosawa H, et al.
       dopaminergic neurons in a transgenic mouse model of PD. Cell Death                  Intragenic deletion in the gene encoding ubiquitin carboxy-terminal
       Differ. 2012;19:1623–33.                                                            hydrolase in gad mice. Nat Genet. 1999;23:47–51.
158.   Ramonet D, Daher JPL, Lin BM, Stafa K, Kim J, Banerjee R, et al. Dopa-         180. Setsuie R, Wang Y-L, Mochizuki H, Osaka H, Hayakawa H, Ichihara N,
       minergic neuronal loss, reduced neurite complexity and autophagic                   et al. Dopaminergic neuronal loss in transgenic mice expressing the
       abnormalities in transgenic mice expressing G2019S mutant LRRK2.                    Parkinson’s disease-associated UCH-L1 I93M mutant. Neurochem Int.
       PLoS ONE. 2011;6:e18568.                                                            2007;50:119–29.
159.   Kozina E, Sadasivan S, Jiao Y, Dou Y, Ma Z, Tan H, et al. Mutant LRRK2         181. Tran HH, Dang SNA, Nguyen TT, Huynh AM, Dao LinhM, Kamei K,
       mediates peripheral and central immune responses leading to neuro-                  et al. Drosophila ubiquitin c-terminal hydrolase knockdown model of
       degeneration in vivo. Brain J Neurol. 2018;141:1753–69.                             Parkinson’s disease. Sci Rep. 2018;8:4468.
160.   Xiong Y, Neifert S, Karuppagounder SS, Liu Q, Stankowski JN, Lee BD,           182. O’Regan G, deSouza R-M, Balestrino R, Schapira AH. Glucocerebrosi-
       et al. Robust kinase- and age-dependent dopaminergic and norepi-                    dase mutations in Parkinson disease. J Park Dis. 2017;7:411–22.
       nephrine neurodegeneration in LRRK2 G2019S transgenic mice. Proc               183. Gan-Or Z, Liong C, Alcalay RN. GBA-associated Parkinson’s disease
       Natl Acad Sci U S A. 2018;115:1635–40.                                              and other synucleinopathies. Curr Neurol Neurosci Rep. 2018;18:44.
161.   Nagakubo D, Taira T, Kitaura H, Ikeda M, Tamai K, Iguchi-Ariga SMM,            184. Winfield SL, Tayebi N, Martin BM, Ginns EI, Sidransky E. Identification
       et al. DJ-1, a novel oncogene which transforms mouse NIH3T3 cells in                of three additional genes contiguous to the glucocerebrosidase
       cooperation withras. Biochem Biophys Res Commun. 1997;231:509–13.                   locus on chromosome 1q21: implications for Gaucher disease.
162.   Ariga H, Takahashi-Niki K, Kato I, Maita H, Niki T, Iguchi-Ariga SMM.               Genome Res. 1997;7:1020–6.
       Neuroprotective function of DJ-1 in Parkinson’s disease. Oxid Med Cell         185. Taguchi YV, Liu J, Ruan J, Pacheco J, Zhang X, Abbasi J, et al. Glucosyl-
       Longev. 2013;2013:1–9.                                                              sphingosine promotes α-synuclein pathology in mutant GBA-associ-
163.   Aleyasin H, Rousseaux MWC, Marcogliese PC, Hewitt SJ, Irrcher I,                    ated Parkinson’s disease. J Neurosci. 2017;37:9617–31.
       Joselin AP, et al. DJ-1 protects the nigrostriatal axis from the neurotoxin    186. Blauwendraat C, Reed X, Krohn L, Heilbron K, Bandres-Ciga S, Tan
       MPTP by modulation of the AKT pathway. Proc Natl Acad Sci U S A.                    M, et al. Genetic modifiers of risk and age at onset in GBA associ-
       2010;107:3186–91.                                                                   ated Parkinson’s disease and Lewy body dementia. Brain J Neurol.
164.   Trempe J-F, Fon EA. Structure and function of parkin, PINK1, and DJ-1,              2020;143:234–48.
       the three musketeers of neuroprotection. Front Neurol. 2013;4.                 187. Avenali M, Blandini F, Cerri S. Glucocerebrosidase defects as a major
165.   Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, et al.                 risk factor for Parkinson’s disease. Front Aging Neurosci. 2020;12:97.
       Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-          188. Behl T, Kaur G, Fratila O, Buhas C, Judea-Pusta CT, Negrut N, et al.
       tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci U S             Cross-talks among GBA mutations, glucocerebrosidase, and
       A. 2005;102:5215–20.                                                                α-synuclein in GBA-associated Parkinson’s disease and their targeted
166.   Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, et al.        therapeutic approaches: a comprehensive review. Transl Neurode-
       Mutations in the DJ-1 gene associated with autosomal recessive early-               gener. 2021;10:4.
       onset parkinsonism. Science. 2003;299:256–9.                                   189. Goldin E. Gaucher disease and parkinsonism, a molecular link theory.
167.   Xu C-Y, Kang W-Y, Chen Y-M, Jiang T-F, Zhang J, Zhang L-N, et al. DJ-1              Mol Genet Metab. 2010;101:307–10.
       inhibits α-synuclein aggregation by regulating chaperone-mediated              190. Migdalska-Richards A, Schapira AHV. The relationship between
       autophagy. Front Aging Neurosci. 2017;9:308.                                        glucocerebrosidase mutations and Parkinson disease. J Neurochem.
168.   Lopert P, Patel M. Brain mitochondria from DJ-1 knockout mice show                  2016;139(Suppl 1):77–90.
       increased respiration-dependent hydrogen peroxide consumption.                 191. Farfel-Becker T, Do J, Tayebi N, Sidransky E. Can GBA1-associated
       Redox Biol. 2014;2:667–72.                                                          Parkinson disease be modeled in the mouse? Trends Neurosci.
169.   Chen L, Cagniard B, Mathews T, Jones S, Koh HC, Ding Y, et al. Age-                 2019;42:631–43.
       dependent motor deficits and dopaminergic dysfunction in DJ-1 null             192. Ginns EI, Mak SK-K, Ko N, Karlgren J, Akbarian S, Chou VP, et al. Neuro-
       mice. J Biol Chem. 2005;280:21418–26.                                               inflammation and α-synuclein accumulation in response to glucocer-
170.   Aryal B, Lee Y. Disease model organism for Parkinson disease: Drosophila            ebrosidase deficiency are accompanied by synaptic dysfunction. Mol
       melanogaster. BMB Rep. 2019;52:250–8.                                               Genet Metab. 2014;111:152–62.
171.   Ji Y-J, Wang H-L, Yin B-L, Ren X-Y. Down-regulation of DJ-1 augments           193. Li H, Ham A, Ma TC, Kuo S-H, Kanter E, Kim D, et al. Mitochondrial
       neuroinflammation via Nrf2/Trx1/NLRP3 axis in MPTP-induced Parkin-                  dysfunction and mitophagy defect triggered by heterozygous GBA
       son’s disease mouse model. Neuroscience. 2020;442:253–63.                           mutations. Autophagy. 2019;15:113–30.
172.   Wilkinson KD, Lee KM, Deshpande S, Duerksen-Hughes P, Boss JM, Pohl            194. Migdalska-Richards A, Wegrzynowicz M, Rusconi R, Deangeli G, Di
       J. The neuron-specific protein PGP 9.5 is a ubiquitin carboxyl-terminal             Monte DA, Spillantini MG, et al. The L444P Gba1 mutation enhances
       hydrolase. Science. 1989;246:670–3.
Dovonou et al. Translational Neurodegeneration             (2023) 12:36                                                                            Page 22 of 25
       alpha-synuclein induced loss of nigral dopaminergic neurons in               216. Zou HL, Su CJ, Shi M, Zhao GY, Li ZY, Guo C, et al. Expression of the LIM-
       mice. Brain J Neurol. 2017;140:2706–21.                                           homeodomain gene Lmx1a in the postnatal mouse central nervous
195.   Abul Khair SB, Dhanushkodi NR, Ardah MT, Chen W, Yang Y, Haque                    system. Brain Res Bull. 2009;78:306–12.
       ME. Silencing of glucocerebrosidase gene in Drosophila enhances              217. Doucet-Beaupré H, Gilbert C, Profes MS, Chabrat A, Pacelli C, Giguère N,
       the aggregation of Parkinson’s disease associated α-synuclein mutant              et al. Lmx1a and Lmx1b regulate mitochondrial functions and survival
       A53T and affects locomotor activity. Front Neurosci. 2018;12:81.                  of adult midbrain dopaminergic neurons. Proc Natl Acad Sci U S A.
196.   Maor G, Cabasso O, Krivoruk O, Rodriguez J, Steller H, Segal D, et al.            2016;113:E4387–4396.
       The contribution of mutant GBA to the development of Parkinson               218. Borgkvist A, Puelles E, Carta M, Acampora D, Ang S-L, Wurst W, et al.
       disease in Drosophila. Hum Mol Genet. 2016;25:2712–27.                            Altered dopaminergic innervation and amphetamine response in adult
197.   Mazzulli JR, Xu Y-H, Sun Y, Knight AL, McLean PJ, Caldwell GA, et al.             Otx2 conditional mutant mice. Mol Cell Neurosci. 2006;31:293–302.
       Gaucher disease glucocerebrosidase and α-synuclein form a bidirec-           219. Chung CY, Licznerski P, Alavian KN, Simeone A, Lin Z, Martin E, et al.
       tional pathogenic loop in synucleinopathies. Cell. 2011;146:37–52.                The transcription factor orthodenticle homeobox 2 influences axonal
198.   Do J, McKinney C, Sharma P, Sidransky E. Glucocerebrosidase and its               projections and vulnerability of midbrain dopaminergic neurons. Brain
       relevance to Parkinson disease. Mol Neurodegener. 2019;14:36.                     J Neurol. 2010;133:2022–31.
199.   Blaess S, Ang S-L. Genetic control of midbrain dopaminergic neuron           220. Domanskyi A, Alter H, Vogt MA, Gass P, Vinnikov IA. Transcription factors
       development. Wiley Interdiscip Rev Dev Biol. 2015;4:113–34.                       Foxa1 and Foxa2 are required for adult dopamine neurons mainte-
200.   Brodski C, Blaess S, Partanen J, Prakash N. Crosstalk of intercellular            nance. Front Cell Neurosci. 2014;8:275.
       signaling pathways in the generation of midbrain dopaminergic                221. Hwang D-Y, Ardayfio P, Kang UJ, Semina EV, Kim K-S. Selective loss of
       neurons in vivo and from stem cells. J Dev Biol. 2019;7:3.                        dopaminergic neurons in the substantia nigra of Pitx3-deficient aphakia
201.   Alavian KN, Jeddi S, Naghipour SI, Nabili P, Licznerski P, Tierney TS. The        mice. Brain Res Mol Brain Res. 2003;114:123–31.
       lifelong maintenance of mesencephalic dopaminergic neurons by                222. Stott SRW, Metzakopian E, Lin W, Kaestner KH, Hen R, Ang SL. Foxa1
       Nurr1 and engrailed. J Biomed Sci. 2014;21:27.                                    and foxa2 are required for the maintenance of dopaminergic proper-
202.   Alavian KN, Scholz C, Simon HH. Transcriptional regulation of mesen-              ties in ventral midbrain neurons at late embryonic stages. J Neurosci.
       cephalic dopaminergic neurons: the full circle of life and death. Mov             2013;33:8022–34.
       Disord. 2008;23:319–28.                                                      223. van den Munckhof P, Luk KC, Ste-Marie L, Montgomery J, Blanchet PJ,
203.   Doucet-Beaupré H, Ang S-L, Lévesque M. Cell fate determination, neu-              Sadikot AF, et al. Pitx3 is required for motor activity and for survival
       ronal maintenance and disease state: the emerging role of transcription           of a subset of midbrain dopaminergic neurons. Dev Camb Engl.
       factors Lmx1a and Lmx1b. FEBS Lett. 2015;589:3727–38.                             2003;130:2535–42.
204.   Fuchs J, Mueller JC, Lichtner P, Schulte C, Munz M, Berg D, et al. The       224. Kadkhodaei B, Ito T, Joodmardi E, Mattsson B, Rouillard C, Carta M, et al.
       transcription factor PITX3 is associated with sporadic Parkinson’s dis-           Nurr1 is required for maintenance of maturing and adult midbrain
       ease. Neurobiol Aging. 2009;30:731–8.                                             dopamine neurons. J Neurosci. 2009;29:15923–32.
205.   Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S,           225. Nordström U, Beauvais G, Ghosh A, Pulikkaparambil Sasidharan BC,
       Chang D, et al. Identification of novel risk loci, causal insights, and           Lundblad M, Fuchs J, et al. Progressive nigrostriatal terminal dysfunction
       heritable risk for Parkinson’s disease: a meta-analysis of genome-wide            and degeneration in the engrailed1 heterozygous mouse model of
       association studies. Lancet Neurol. 2019;18:1091–102.                             Parkinson’s disease. Neurobiol Dis. 2015;73:70–82.
206.   Bergman O, Håkansson A, Westberg L, Belin AC, Sydow O, Olson L, et al.       226. Kadkhodaei B, Alvarsson A, Schintu N, Ramsköld D, Volakakis N, Jood-
       Do polymorphisms in transcription factors LMX1A and LMX1B influ-                  mardi E, et al. Transcription factor Nurr1 maintains fiber integrity and
       ence the risk for Parkinson’s disease? J Neural Transm Vienna Austria             nuclear-encoded mitochondrial gene expression in dopamine neurons.
       1996. 2009;116:333–8.                                                             Proc Natl Acad Sci U S A. 2013;110:2360–5.
207.   Bergman O, Westberg L, Nilsson L-G, Adolfsson R, Eriksson E. Preliminary     227. Wurst W, Auerbach AB, Joyner AL. Multiple developmental defects
       evidence that polymorphisms in dopamine-related transcription fac-                in Engrailed-1 mutant mice: an early mid-hindbrain deletion and
       tors LMX1A, LMX1B and PITX3 are associated with schizophrenia. Prog               patterning defects in forelimbs and sternum. Dev Camb Engl.
       Neuropsychopharmacol Biol Psychiatry. 2010;34:1094–7.                             1994;120:2065–75.
208.   Haubenberger D, Reinthaler E, Mueller JC, Pirker W, Katzenschlager           228. Baiguera C, Alghisi M, Pinna A, Bellucci A, De Luca MA, Frau L, et al.
       R, Froehlich R, et al. Association of transcription factor polymor-               Late-onset parkinsonism in NFκB/c-Rel-deficient mice. Brain J Neurol.
       phisms PITX3 and EN1 with Parkinson’s disease. Neurobiol Aging.                   2012;135:2750–65.
       2011;32:302–7.                                                               229. Giguère N, Burke Nanni S, Trudeau LE. On cell loss and selective vulner-
209.   Laguna A, Schintu N, Nobre A, Alvarsson A, Volakakis N, Jacobsen JK,              ability of neuronal populations in Parkinson’s disease. Front Neurol.
       et al. Dopaminergic control of autophagic-lysosomal function impli-               2018;9:455.
       cates Lmx1b in Parkinson’s disease. Nat Neurosci. 2015;18:826–35.            230. Surmeier DJ. Determinants of dopaminergic neuron loss in Parkinson’s
210.   Chabrat A, Brisson G, Doucet-Beaupré H, Salesse C, Schaan Profes M,               disease. FEBS J. 2018;285:3657–68.
       Dovonou A, et al. Transcriptional repression of Plxnc1 by Lmx1a and          231. Haelterman NA, Yoon WH, Sandoval H, Jaiswal M, Shulman JM, Bellen
       Lmx1b directs topographic dopaminergic circuit formation. Nat Com-                HJ. A mitocentric view of Parkinson’s disease. Annu Rev Neurosci.
       mun. 2017;8:933.                                                                  2014;37:137–59.
211.   Deng Q, Andersson E, Hedlund E, Alekseenko Z, Coppola E, Panman              232. Piccinin E, Sardanelli AM, Seibel P, Moschetta A, Cocco T, Villani G. PGC-
       L, et al. Specific and integrated roles of Lmx1a, Lmx1b and Phox2a in             1s in the spotlight with Parkinson’s disease. Int J Mol Sci. 2021;22:3487.
       ventral midbrain development. Dev Camb Engl. 2011;138:3399–408.              233. Faustini G, Bono F, Valerio A, Pizzi M, Spano P, Bellucci A. Mitochondria
212.   Salesse C, Charest J, Doucet-Beaupré H, Castonguay A-M, Labrecque S,              and α-synuclein: friends or foes in the pathogenesis of Parkinson’s
       De Koninck P, et al. Opposite control of excitatory and inhibitory syn-           disease? Genes. 2017;8:E377.
       apse formation by Slitrk2 and Slitrk5 on dopamine neurons modulates          234. Schapira AHV, Cooper JM, Dexter D, Jenner P, Clark JB, Marsden CD.
       hyperactivity behavior. Cell Rep. 2020;30:2374–2386e5.                            Mitochondrial complex I deficiency in Parkinson’s disease. Lancet.
213.   Yan CH, Levesque M, Claxton S, Johnson RL, Ang SL. Lmx1a and lmx1b                1989;333:1269.
       function cooperatively to regulate proliferation, specification, and         235. Hauser DN, Primiani CT, Langston RG, Kumaran R, Cookson MR. The
       differentiation of midbrain dopaminergic progenitors. J Neurosci.                 Polg mutator phenotype does not cause dopaminergic neurodegen-
       2011;31:12413–25.                                                                 eration in DJ-1-deficient mice. eNeuro. 2015;2:ENEURO.0075-14.2015.
214.   Asbreuk C. CNS expression pattern of Lmx1b and coexpression with             236. Scott L, Karuppagounder SS, Neifert S, Kang BG, Wang H, Dawson VL,
       ptx genes suggest functional cooperativity in the development of                  et al. The absence of parkin does not promote dopamine or mitochon-
       forebrain motor control systems. Mol Cell Neurosci. 2002;21:410–20.               drial dysfunction in PolgAD257A/D257A mitochondrial mutator mice. J
215.   Dai J-X, Hu Z-L, Shi M, Guo C, Ding Y-Q. Postnatal ontogeny of the tran-          Neurosci. 2022;42:9263–77.
       scription factor Lmx1b in the mouse central nervous system. J Comp
       Neurol. 2008;509:341–55.
Dovonou et al. Translational Neurodegeneration             (2023) 12:36                                                                              Page 23 of 25
237. Kim H-W, Choi W-S, Sorscher N, Park HJ, Tronche F, Palmiter RD, et al.         258. Ruiz R, Biea IA, Tabares L, α-Synuclein A30P decreases neurodegenera-
     Genetic reduction of mitochondrial complex I function does not lead                 tion and increases synaptic vesicle release probability in CSPα-null
     to loss of dopamine neurons in vivo. Neurobiol Aging. 2015;36:2617–27.              mice. Neuropharmacology. 2014;76:106–17.
238. González-Rodríguez P, Zampese E, Stout KA, Guzman JN, Ilijic E, Yang           259. Chen AY, Wilburn P, Hao X, Tully T. Walking deficits and centrophobism
     B, et al. Disruption of mitochondrial complex I induces progressive                 in an α-synuclein fly model of Parkinson’s disease. Genes Brain Behav.
     parkinsonism. Nature. 2021;599:650–6.                                               2014;13:812–20.
239. Bartels T, Choi JG, Selkoe DJ. α-Synuclein occurs physiologically as a hel-    260. Mizuno H, Fujikake N, Wada K, Nagai Y. α-Synuclein transgenic dros-
     ically folded tetramer that resists aggregation. Nature. 2011;477:107–10.           ophila as a model of Parkinson’s disease and related synucleinopathies.
240. Burré J, Sharma M, Südhof TC. Cell biology and pathophysiology of                   Park Dis. 2010;2011:212706.
     α-synuclein. Cold Spring Harb Perspect Med. 2018;8.                            261. Perni M, van der Goot A, Limbocker R, van Ham TJ, Aprile FA, Xu CK
241. Gómez-Benito M, Granado N, García-Sanz P, Michel A, Dumoulin M,                     et al. Comparative studies in the A30P and A53T α-synuclein C. elegans
     Moratalla R. Modeling Parkinson’s disease with the alpha-synuclein                  strains to investigate the molecular origins of Parkinson’s disease. Front
     protein. Front Pharmacol. 2020;11:356.                                              Cell Dev Biol. 2021;9.
242. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. Alpha-            262. Kuwahara T, Koyama A, Koyama S, Yoshina S, Ren C-H, Kato T, et al. A
     synuclein in filamentous inclusions of Lewy bodies from Parkinson’s                 systematic RNAi screen reveals involvement of endocytic pathway in
     disease and dementia with lewy bodies. Proc Natl Acad Sci U S A.                    neuronal dysfunction in α-synuclein transgenic C. elegans. Hum Mol
     1998;95:6469–73.                                                                    Genet. 2008;17:2997–3009.
243. Huang M, Wang B, Li X, Fu C, Wang C, Kang X. α-Synuclein: a multifunc-         263. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A,
     tional player in exocytosis, endocytosis, and vesicle recycling. Front              et al. Mutation in the alpha-synuclein gene identified in families with
     Neurosci. 2019;13:28.                                                               Parkinson’s disease. Science. 1997;276:2045–7.
244. Liu H, Koros C, Strohäker T, Schulte C, Bozi M, Varvaresos S, et al. A novel   264. Giasson BI, Duda JE, Quinn SM, Zhang B, Trojanowski JQ, Lee VM-Y.
     SNCA A30G mutation causes familial Parkinsonʼs disease. Mov Disord.                 Neuronal α-synucleinopathy with severe movement disorder in mice
     2021;36:1624–33.                                                                    expressing A53T human α-synuclein. Neuron. 2002;34:521–33.
245. Oczkowska A, Kozubski W, Lianeri M, Dorszewska J. Mutations in PRKN            265. Mougenot A-LJ, Bencsik A, Nicot S, Vulin J, Morignat E, Verchère J, et al.
     and SNCA genes important for the progress of Parkinson’s disease. Curr              Transmission of prion strains in a transgenic mouse model overexpress-
     Genomics. 2013;14:502–17.                                                           ing human A53T mutated α-synuclein. J Neuropathol Exp Neurol.
246. Rockenstein E, Mallory M, Hashimoto M, Song D, Shults CW, Lang I, et al.            2011;70:377–85.
     Differential neuropathological alterations in transgenic mice expressing       266. Teravskis PJ, Covelo A, Miller EC, Singh B, Martell-Martínez HA, Benney-
     alpha-synuclein from the platelet-derived growth factor and Thy-1                   worth MA, et al. A53T mutant alpha-synuclein induces tau-dependent
     promoters. J Neurosci Res. 2002;68:568–78.                                          postsynaptic impairment independently of neurodegenerative
247. Subramaniam SR, Vergnes L, Franich NR, Reue K, Chesselet M-F. Region                changes. J Neurosci. 2018;38:9754–67.
     specific mitochondrial impairment in mice with widespread overex-              267. Golbe LI, Lazzarini AM, Duvoisin RC, Iorio GD, Sanges G, Bonavita V, et al.
     pression of alpha-synuclein. Neurobiol Dis. 2014;70:204–13.                         Clinical genetic analysis of Parkinson’s disease in the contursi kindred.
248. Watson MB, Richter F, Lee SK, Gabby L, Wu J, Masliah E, et al. Regionally-          Ann Neurol. 1996;40:767–75.
     specific microglial activation in young mice over-expressing human             268. Ramsey CP, Tsika E, Ischiropoulos H, Giasson BI. DJ-1 deficient mice
     wildtype alpha-synuclein. Exp Neurol. 2012;237:318–34.                              demonstrate similar vulnerability to pathogenic Ala53Thr human -syn
249. Chesselet M-F, Richter F, Zhu C, Magen I, Watson MB, Subramaniam SR.                toxicity. Hum Mol Genet. 2010;19:1425–37.
     A progressive mouse model of Parkinson’s disease: the Thy1-aSyn (“Line         269. Macdonald JA, Chen JL, Masuda-Suzukake M, Schweighauser M,
     61”) mice. Neurother J Am Soc Exp Neurother. 2012;9:297–314.                        Jaunmuktane Z, Warner T, et al. Assembly of α-synuclein and neurode-
250. Wegrzynowicz M, Bar-On D, Calo’ L, Anichtchik O, Iovino M, Xia J, et al.            generation in the central nervous system of heterozygous M83 mice
     Depopulation of dense α-synuclein aggregates is associated with res-                following the peripheral administration of α-synuclein seeds. Acta
     cue of dopamine neuron dysfunction and death in a new Parkinson’s                   Neuropathol Commun. 2021;9:189.
     disease model. Acta Neuropathol (Berl). 2019;138:575–95.                       270. Sorrentino ZA, Xia Y, Funk C, Riffe CJ, Rutherford NJ, Ceballos Diaz
251. Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Ampuero I,                C, et al. Motor Neuron loss and neuroinflammation in a model
     et al. The new mutation, E46K, of α-synuclein causes parkinson and                  of α-Synuclein-induced neurodegeneration. Neurobiol Dis.
     Lewy body dementia: New α-Synuclein gene mutation. Ann Neurol.                      2018;120:98–106.
     2004;55:164–73.                                                                271. Martin LJ, Pan Y, Price AC, Sterling W, Copeland NG, Jenkins NA, et al.
252. Emmer KL, Waxman EA, Covy JP, Giasson BI. E46K human α-synuclein                    Parkinson’s disease alpha-synuclein transgenic mice develop neuronal
     transgenic mice develop Lewy-like and Tau pathology associated                      mitochondrial degeneration and cell death. J Neurosci. 2006;26:41–50.
     with age-dependent, detrimental motor impairment. J Biol Chem.                 272. Malkus KA, Ischiropoulos H. Regional deficiencies in chaperone-medi-
     2011;286:35104–18.                                                                  ated autophagy underlie α-synuclein aggregation and neurodegenera-
253. Cannon JR, Geghman KD, Tapias V, Sew T, Dail MK, Li C, et al. Expression            tion. Neurobiol Dis. 2012;46:732–44.
     of human E46K-mutated α-synuclein in BAC-transgenic rats replicates            273. Chung HK, Ho H-A, Pérez-Acuña D, Lee S-J. Modeling α-Synuclein prop-
     early-stage Parkinson’s disease features and enhances vulnerability to              agation with preformed Fibril injections. J Mov Disord. 2019;12:139–51.
     mitochondrial impairment. Exp Neurol. 2013;240:44–56.                          274. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, et al. Patho-
254. Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, Kösel S, et al. AlaSO-           logical α-synuclein transmission initiates Parkinson-like neurodegenera-
     Pro mutation in the gene encoding α-synuclein in Parkinson’s disease.               tion in nontransgenic mice. Science. 2012;338:949–53.
     Nat Genet. 1998;18:106–8.                                                      275. Sargent D, Verchère J, Lazizzera C, Gaillard D, Lakhdar L, Streichenberger
255. Piltonen M, Savolainen M, Patrikainen S, Baekelandt V, Myöhänen TT,                 N, et al. Prion-like’ propagation of the synucleinopathy of M83 trans-
     Männistö PT. Comparison of motor performance, brain biochemistry                    genic mice depends on the mouse genotype and type of inoculum. J
     and histology of two A30P α-synuclein transgenic mouse strains. Neu-                Neurochem. 2017;143:126–35.
     roscience. 2013;231:157–68.                                                    276. Bétemps D, Verchère J, Brot S, Morignat E, Bousset L, Gaillard D, et al.
256. Ekmark-Lewén S, Lindström V, Gumucio A, Ihse E, Behere A, Kahle PJ,                 Alpha-synuclein spreading in M83 mice brain revealed by detection of
     et al. Early fine motor impairment and behavioral dysfunction in (Thy-              pathological α-synuclein by enhanced ELISA. Acta Neuropathol Com-
     1)-h[A30P] alpha-synuclein mice. Brain Behav. 2018;8:e00915.                        mun. 2014;2:29.
257. Poon HF, Frasier M, Shreve N, Calabrese V, Wolozin B, Butterfield DA.          277. Luk KC, Kehm VM, Zhang B, O’Brien P, Trojanowski JQ, Lee VMY.
     Mitochondrial associated metabolic proteins are selectively oxidized in             Intracerebral inoculation of pathological α-synuclein initiates a rapidly
     A30P alpha-synuclein transgenic mice–a model of familial Parkinson’s                progressive neurodegenerative α-synucleinopathy in mice. J Exp Med.
     disease. Neurobiol Dis. 2005;18:492–8.                                              2012;209:975–86.
                                                                                    278. Sorrentino ZA, Xia Y, Funk C, Riffe CJ, Rutherford NJ, Cebal-
                                                                                         los Diaz C, et al. Motor neuron loss and neuroinflammation in a
Dovonou et al. Translational Neurodegeneration                (2023) 12:36                                                                                   Page 24 of 25
       model of α-synuclein-induced neurodegeneration. Neurobiol Dis.                   300. Ip CW, Klaus L-C, Karikari AA, Visanji NP, Brotchie JM, Lang AE, et al.
       2018;120:98–106.                                                                      AAV1/2-induced overexpression of A53T-α-synuclein in the substantia
279.   Recasens A, Dehay B, Bové J, Carballo-Carbajal I, Dovero S, Pérez-Villalba            nigra results in degeneration of the nigrostriatal system with Lewy-like
       A, et al. Lewy body extracts from Parkinson disease brains trigger                    pathology and motor impairment: a new mouse model for Parkinson’s
       α-synuclein pathology and neurodegeneration in mice and monkeys.                      disease. Acta Neuropathol Commun. 2017;5:11.
       Ann Neurol. 2014;75:351–62.                                                      301. Van der Perren A, Van den Haute C, Baekelandt V. Viral vector-
280.   Okuda S, Uemura N, Sawamura M, Taguchi T, Ikuno M, Uemura MT, et al.                  based models of Parkinson’s disease. Curr Top Behav Neurosci.
       Rapid induction of dopaminergic neuron loss accompanied by Lewy                       2015;22:271–301.
       body-like inclusions in A53T BAC-SNCA transgenic mice. Neurothera-               302. Recasens A, Dehay B. Alpha-synuclein spreading in Parkinson’s disease.
       peutics. 2022;19(1):289–304.                                                          Front Neuroanat. 2014;8:159.
281.   Wang X, Becker K, Levine N, Zhang M, Lieberman AP, Moore DJ, et al.              303. Koprich JB, Johnston TH, Reyes MG, Sun X, Brotchie JM. Expression of
       Pathogenic alpha-synuclein aggregates preferentially bind to mito-                    human A53T alpha-synuclein in the rat substantia nigra using a novel
       chondria and affect cellular respiration. Acta Neuropathol Commun.                    AAV1/2 vector produces a rapidly evolving pathology with protein
       2019;7:41.                                                                            aggregation, dystrophic neurite architecture and nigrostriatal degen-
282.   Guiney SJ, Adlard PA, Lei P, Mawal CH, Bush AI, Finkelstein DI, et al. Fibril-        eration with potential to model the pathology of Parkinson’s disease.
       lar α-synuclein toxicity depends on functional lysosomes. J Biol Chem.                Mol Neurodegener. 2010;5:43.
       2020;295:17497–513.                                                              304. Koprich JB, Johnston TH, Huot P, Reyes MG, Espinosa M, Brotchie JM.
283.   Masuda-Suzukake M, Nonaka T, Hosokawa M, Oikawa T, Arai T, Akiyama                    Progressive neurodegeneration or endogenous compensation in an
       H, et al. Prion-like spreading of pathological α-synuclein in brain. Brain.           animal model of Parkinson’s disease produced by decreasing doses of
       2013;136:1128–38.                                                                     alpha-synuclein. PLoS ONE. 2011;6:e17698.
284.   Polinski NK. A summary of phenotypes observed in the in vivo rodent              305. Dehay B, Fernagut P-O. Alpha-synuclein-based models of Parkinson’s
       alpha-synuclein preformed fibril model. J Park Dis. 2021;11:1555–67.                  disease. Rev Neurol (Paris). 2016;172:371–8.
285.   Ahn EH, Kang SS, Liu X, Chen G, Zhang Z, Chandrasekharan B, et al.               306. Huntington TE, Srinivasan R. Adeno-associated virus expression of
       Initiation of Parkinson’s disease from gut to brain by δ-secretase. Cell              α-synuclein as a tool to model Parkinson’s disease: current understand-
       Res. 2020;30:70–87.                                                                   ing and knowledge gaps. Aging Dis. 2021;12:1120–37.
286.   Challis C, Hori A, Sampson TR, Yoo BB, Challis RC, Hamilton AM, et al.           307. Volpicelli-Daley LA, Kirik D, Stoyka LE, Standaert DG, Harms AS. How
       Gut-seeded α-synuclein fibrils promote gut dysfunction and brain                      can rAAV-α-synuclein and the fibril α-synuclein models advance our
       pathology specifically in aged mice. Nat Neurosci. 2020;23:327–36.                    understanding of Parkinson’s disease? J Neurochem. 2016;139:131–55.
287.   Kim S, Kwon S-H, Kam T-I, Panicker N, Karuppagounder SS, Lee S, et al.           308. Decressac M, Mattsson B, Lundblad M, Weikop P, Björklund A. Pro-
       Transneuronal propagation of pathologic α-synuclein from the gut to                   gressive neurodegenerative and behavioural changes induced by
       the brain models Parkinson’s disease. Neuron. 2019;103:627–641e7.                     AAV-mediated overexpression of α-synuclein in midbrain dopamine
288.   Earls RH, Menees KB, Chung J, Barber J, Gutekunst C-A, Hazim MG, et al.               neurons. Neurobiol Dis. 2012;45:939–53.
       Intrastriatal injection of preformed alpha-synuclein fibrils alters central      309. Kirik D, Annett LE, Burger C, Muzyczka N, Mandel RJ, Björklund A.
       and peripheral immune cell profiles in non-transgenic mice. J Neuroin-                Nigrostriatal alpha-synucleinopathy induced by viral vector-mediated
       flammation. 2019;16:250.                                                              overexpression of human alpha-synuclein: a new primate model of
289.   Mahul-Mellier A-L, Burtscher J, Maharjan N, Weerens L, Croisier M,                    Parkinson’s disease. Proc Natl Acad Sci U S A. 2003;100:2884–9.
       Kuttler F, et al. The process of Lewy body formation, rather than simply         310. Eslamboli A, Romero-Ramos M, Burger C, Bjorklund T, Muzyczka N, Man-
       α-synuclein fibrillization, is one of the major drivers of neurodegenera-             del RJ, et al. Long-term consequences of human alpha-synuclein over-
       tion. Proc Natl Acad Sci U S A. 2020;117:4971–82.                                     expression in the primate ventral midbrain. Brain. 2007;130:799–815.
290.   Polinski NK, Volpicelli-Daley LA, Sortwell CE, Luk KC, Cremades N, Got-          311. Yang W, Wang G, Wang C-E, Guo X, Yin P, Gao J, et al. Mutant alpha-
       tler LM, et al. Best practices for generating and using alpha-synuclein               synuclein causes age-dependent neuropathology in monkey brain. J
       pre-formed fibrils to model Parkinson’s disease in rodents. J Park Dis.               Neurosci. 2015;35:8345–58.
       2018;8:303–22.                                                                   312. Di Maio R, Barrett PJ, Hoffman EK, Barrett CW, Zharikov A, Borah A, et al.
291.   van Ham TJ, Thijssen KL, Breitling R, Hofstra RMW, Plasterk RHA, Nollen               α-Synuclein binds to TOM20 and inhibits mitochondrial protein import
       EAA. C. elegans model identifies genetic modifiers of α-synuclein inclu-              in Parkinson’s disease. Sci Transl Med. 2016. https://doi.org/10.1126/scitr
       sion formation during aging. PLoS Genet. 2008;4:e1000027.                             anslmed.aaf3634.
292.   Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, Bhullar B, et al.          313. McKinnon C, De Snoo ML, Gondard E, Neudorfer C, Chau H, Ngana SG,
       Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in               et al. Early-onset impairment of the ubiquitin-proteasome system in
       Parkinson’s models. Science. 2006;313:324–8.                                          dopaminergic neurons caused by α-synuclein. Acta Neuropathol Com-
293.   Cooper JF, Spielbauer KK, Senchuk MM, Nadarajan S, Colaiácovo MP, Van                 mun. 2020;8:17.
       Raamsdonk JM. α-synuclein expression from a single copy transgene                314. Yamada M, Iwatsubo T, Mizuno Y, Mochizuki H. Overexpression of
       increases sensitivity to stress and accelerates neuronal loss in genetic              alpha-synuclein in rat substantia nigra results in loss of dopaminergic
       models of Parkinson’s disease. Exp Neurol. 2018;310:58–69.                            neurons, phosphorylation of alpha-synuclein and activation of cas-
294.   Feany MB, Bender WW. A Drosophila model of Parkinson’s disease.                       pase-9: resemblance to pathogenetic changes in Parkinson’s disease. J
       Nature. 2000;404:394–8.                                                               Neurochem. 2004;91:451–61.
295.   Auluck PK. Chaperone suppression of alpha -synuclein toxicity in a               315. Chung CY, Koprich JB, Siddiqi H, Isacson O. Dynamic changes in presyn-
       Drosophila model for Parkinson’s disease. Science. 2002;295:865–8.                    aptic and axonal transport proteins combined with striatal neuroin-
296.   Ordonez DG, Lee MK, Feany MB. α-synuclein induces mitochondrial                       flammation precede dopaminergic neuronal loss in a rat model of AAV
       dysfunction through spectrin and the actin cytoskeleton. Neuron.                      alpha-synucleinopathy. J Neurosci. 2009;29:3365–73.
       2018;97:108–124e6.                                                               316. Sanchez-Guajardo V, Febbraro F, Kirik D, Romero-Ramos M. Micro-
297.   Lu J, Sun F, Ma H, Qing H, Deng Y. Comparison between α-synuclein                     glia acquire distinct activation profiles depending on the degree of
       wild-type and A53T mutation in a progressive Parkinson’s disease                      alpha-synuclein neuropathology in a rAAV based model of Parkinson’s
       model. Biochem Biophys Res Commun. 2015;464:988–93.                                   disease. PLoS ONE. 2010;5:e8784.
298.   Lo Bianco C, Ridet J-L, Schneider BL, Deglon N, Aebischer P. Alpha-              317. Theodore S, Cao S, McLean PJ, Standaert DG. Targeted overexpres-
       synucleinopathy and selective dopaminergic neuron loss in a rat                       sion of human alpha-synuclein triggers microglial activation and an
       lentiviral-based model of Parkinson’s disease. Proc Natl Acad Sci U S A.              adaptive immune response in a mouse model of Parkinson disease. J
       2002;99:10813–8.                                                                      Neuropathol Exp Neurol. 2008;67:1149–58.
299.   Kirik D, Rosenblad C, Burger C, Lundberg C, Johansen TE, Muzyczka                318. Ulusoy A, Decressac M, Kirik D, Björklund A. Viral vector-mediated over-
       N, et al. Parkinson-like neurodegeneration induced by targeted over-                  expression of α-synuclein as a progressive model of Parkinson’s disease.
       expression of alpha-synuclein in the nigrostriatal system. J Neurosci.                Prog Brain Res. 2010;184:89–111.
       2002;22:2780–91.
Dovonou et al. Translational Neurodegeneration            (2023) 12:36                                                                            Page 25 of 25